US8937068B2 - Pyridopyrazine derivatives and their use - Google Patents

Pyridopyrazine derivatives and their use Download PDF

Info

Publication number
US8937068B2
US8937068B2 US11/558,493 US55849306A US8937068B2 US 8937068 B2 US8937068 B2 US 8937068B2 US 55849306 A US55849306 A US 55849306A US 8937068 B2 US8937068 B2 US 8937068B2
Authority
US
United States
Prior art keywords
alkyl
aryl
heteroaryl
pyrido
pyrazin
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Fee Related, expires
Application number
US11/558,493
Other versions
US20070123494A1 (en
Inventor
Irene Seipelt
Eckhard Claus
Eckhard Guenther
Tilmann Schuster
Michael Czech
Emmanuel Polymeropoulos
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Aeterna Zentaris GmbH
Original Assignee
Zentaris AG
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Zentaris AG filed Critical Zentaris AG
Priority to US11/558,493 priority Critical patent/US8937068B2/en
Assigned to ZENTARIS GMBH reassignment ZENTARIS GMBH ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: POLYMEROPOULOS, EMMANUEL, SCHUSTER, TILMANN, CLAUS, ECKHARD, CZECH, MICHAEL, SEIPELT, IRENE, GUENTHER, ECKHARD
Publication of US20070123494A1 publication Critical patent/US20070123494A1/en
Application granted granted Critical
Publication of US8937068B2 publication Critical patent/US8937068B2/en
Expired - Fee Related legal-status Critical Current
Adjusted expiration legal-status Critical

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/4985Pyrazines or piperazines ortho- or peri-condensed with heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/08Drugs for disorders of the urinary system of the prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/02Antithrombotic agents; Anticoagulants; Platelet aggregation inhibitors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/02Non-specific cardiovascular stimulants, e.g. drugs for syncope, antihypotensives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems

Definitions

  • the present invention relates to pyridopyrazine derivatives with new biological action and their use for the treatment of physiological and/or pathophysiological states mediated and/or modulated by signal transduction pathways in mammals and in particular in humans.
  • the individual components of these signal cascades are therefore important therapeutic points of attack for intervention in various disease processes (Weinstein-Oppenheimer C. R. et al. 2000, Chang F. et al. 2003, Katso R. et al 2001 and Lu Y. et al 2003).
  • a plurality of growth factors, cytokines and oncogenes transduce their growth-promoting signals via the activation of G-protein coupled ras which leads to the activation of serine threonine kinase Raf and to the activation of mitogen-activated protein kinase kinase 1 and 2 (MAPKK1/2 or Mek1/2) and results in the phosphorylation and activation of MAPK 1 and 2—also known as extracellular signal regulated kinase (Erk1 and 2).
  • the ras-Raf-Mek-Erk signal pathway combines a large number of proto-oncogenes, including ligands, tyrosine kinase receptors, G-proteins, kinases and nuclear transcription factors.
  • Tyrosine kinases such as, for example, EGFR (Mendelsohn J. et al., 2000) frequently mediate constitutively active signals to the downstream ras-Raf-Mek-Erk signal pathway in tumour events caused by overexpression and mutation. Ras mutations are mutated in 30% of all human tumours (Khleif S. N.
  • the members of the Raf kinases also have Mek-Erk-indepedent anti-apoptotic functions whose molecular steps have not yet been fully described.
  • Ask1, Bcl-2, Akt and Bag1 have been described as possible interaction partners for the Mek-Erk-independent Raf activity (Chen J et al., 2001, Troppmaier J. et al., 2003, Rapp U. R. et al., 2004, Gotz R. et al., 2005). It is assumed nowadays that both Mek-Erk-dependent and Mek-Erk-independent signal transduction mechanisms control the activation of the upstream ras and Raf stimuli.
  • the isoenzymes of the phosphatidylinositol 3-kinases function predominantly as lipid kinases and catalyse the D3 phosphorylation of the second-messenger lipids PtdIns (phosphatidylinositol) to PtdIns(3)P, PtdIns(3,4)P 2 , PtdIns(3,4,5)P 3 phosphatidylinositol phosphates.
  • the class I PI3Ks are composed structurally of the catalytic (p110alpha, beta, gamma, delta) and the regulatory (p85alpha, beta or p101gamma) subunits.
  • class II (PI3K-C2alpha, PI3K-C2beta) and class III (Vps34p) enzymes belong to the family of the P13 kinases (Wymann M. P. et al., 1998, VanHaesebroeck B. et al., 2001).
  • the PIP increase triggered by the PI3Ks activates the proliferative ras-Raf-Mek-Erk signal pathway via the coupling of ras on the one hand (Rodriguez-Viciana P.
  • PI3K also have protein-phosphorylating properties (Dhand et al., 1994, Bondeva T. et al., 1998, Bondev A. et al., 1999, VanHaesebroeck B.
  • PI3Ks also have kinase-independent regulating effector properties, e.g. during control of cardiac contraction (Crackower M. A. et al., 2002, Patrucco et al., 2004).
  • PI3Kdelta and PI3Kgamma are specifically expressed on hematopoietic cells and therefore constitute potential points of attack for isoenzyme-specific PI3Kdelta and PI3Kgamma inhibitors in the treatment of inflammatory diseases such as rheumatism, asthma and allergies and in the treatment of B and T cell lymphomas (Okkenhaug K. et al., 2003, Ali K. et al., 2004, Sujobert P. et al., 2005).
  • PI3Kalpha which was recently identified as a proto-oncogene (Shayesteh L. et al., 1999, Ma Y. Y. et al., 2000, Samuels Y.
  • PI3K The kinases related to PI3K (PIKK), which include the serine/threonine kinases mTOR, ATM, ATR, h-SMG-1 and DNA-PK (Chiang G. G. et al 2004) are also of great interest. Their catalytic domains have a high sequence homology to the catalytic domains of PI3Ks.
  • tumour suppressor protein PTEN (Li J. et al., 1997, Steck P. A. et al., 1997)—whose function is the reversion of the phosphorylation initiated by the PI3K—contributes to an overactivation of Akt and its downstream cascade components and thereby emphasise the causal importance of PI3K as a target molecule for tumour therapy.
  • the kinase inhibitor Bay 43-9006 (WO 99/32111, WO 03/068223) which is already undergoing clinical trials, shows a relatively non-specific inhibition pattern of serine/threonine and of tyrosine kinases such as Raf, VEGFR2/3, Flt-3, PDGFR, c-Kit and other kinases.
  • tyrosine kinases such as Raf, VEGFR2/3, Flt-3, PDGFR, c-Kit and other kinases.
  • Angiogenesis-induced advanced tumour diseases e.g. in renal cell carcinoma
  • melanomas having a high B-Raf mutation rate e.g. in melanomas having a high B-Raf mutation rate.
  • the clinical effect of Bay 43-9006 is currently also being determined in patients presenting with refractory solid tumours in combination with, for example, Docetaxel. Mild side effects and promising anti-tumoral effects have been described so far. No inhibition of the
  • the Mek1/2 inhibitor PD0325901 (WO 02/06213) is currently undergoing a Phase I clinical trial.
  • the precursor substance CI-1040 (WO 00/35435, WO 00/37141) was striking because of its high Mek specificity and target affinity.
  • this compound proved to be metabolically unstable in Phase I/II trials.
  • No clinical data are available yet for the current successor substance PD0325901.
  • no interaction with Erk1 or Erk2 nor any PI3K-Akt signal pathway inhibiting function or its simultaneous modulation has yet been disclosed for this Mek inhibitor.
  • Patent specifications WO 04/104002 and WO 04/104003 describe pyrido[2,3-b]pyrazines, which can be substituted in the 6- or 7-position with urea, thiourea, amidine or guanidine groups. These compounds possess properties as inhibitors or modulators of kinases, in particular of tyrosine and serine/threonine kinases, and a use as a medicament is specified. However, no use of these compounds as modulators of lipid kinases, alone or in combination with tyrosine and serine/threonine kinases has been described.
  • patent specification WO 99/17759 describes pyrido[2,3-b]pyrazines which, among other things, carry alkyl-, aryl- and heteroaryl-substituted carbamates in the 6-position. These compounds are to be used to modulate serine threonine protein kinases.
  • Patent specification WO 05/007099 describes, among other things, urea-substituted pyrido[2,3-b]pyrazines as inhibitors of the serine/threonine kinase PKB. A use in the treatment of cancer diseases is specified for these compounds. However, no specific examples of urea-substituted pyridopyrazines with these biological properties are given.
  • pyrido[2,3-b]pyrazines substituted with urea in the 6- and 7-position are given in patent specification WO 05/056547.
  • the compounds in this patent specification are described as inhibitors of protein kinases, in particular GSK-3, Syk and JAK-3. A use in the treatment of proliferative diseases is given for these compounds among other things. No use of these compounds as modulators of lipid kinases, alone or in combination with serine/threonine kinases is described.
  • Patent specification WO 03/024448 describes amide and acrylamide-substituted pyrido[2,3-b]pyrazines which can also contain carbamates as additional substituents and can be used as histone deacetylase inhibitors for the treatment of cell proliferation diseases.
  • PI3K inhibitors published so far are undergoing preclinical trials.
  • ICOS disclosed a PI3K inhibitor IC87114 with high PI3Kdelta isoenzyme specificity (WO 01/81346).
  • Yamanouchi/Piramed describe a selectivity versus the PI3Kalpha isoform.
  • a research environment which has attracted a great deal of attention exists in the early development of PI3K inhibitors (see the review of R. Wetzker et al., 2004).
  • Inhibitors of the SAPK signal pathway either of Jnk or of p38 have been described in the literature (Gum R. J., 1998, Bennett B. L. et al 2001, Davies S. P. et al 2000). However, no PI3K-inhibiting function and also no specific inhibition of Erk1 or Erk2 or simultaneous inhibition of SAPKs, Erk1, Erk2, or PI3Ks is disclosed for these SAPK inhibitors.
  • the object of the present invention is to provide new compounds which can be used for the treatment or prevention of physiological and/or pathophysiological states in mammals, in particular in humans, which are mediated by signal transduction pathways selected from the group consisting of: “ras-Raf-Mek-Erk signal transduction pathway, PI3K-Akt signal transduction pathway and/or SAPK signal transduction pathway”.
  • a further object of the invention is to provide new compounds for the aforesaid uses by modulation of said signal transduction pathways.
  • a further object of the present invention is to provide new compounds which can be used for the treatment or prevention of physiological and/or pathophysiological states in mammals, in particular in humans, which are mediated by enzymes selected from the group consisting of: “ATM, ATR, mTOR, DNA-PK, hSMG-1”.
  • a further object of the invention is to provide new compounds for the aforesaid uses by modulation of said enzymes.
  • R1 and R can be independently of one another (i) hydrogen (ii) hydroxyl (iii) halogen (iv) alkyl, wherein the alkyl group is saturated and can consists of 1 to 8 C atoms, (v) unsubstituted or substituted aryl, wherein the alkyl group can be substituted with one or more, the same or different F, Cl, Br, I, CF 3 , CN, NH 2 , NH-alkyl, NH-cycloalkyl, NH-heterocyclyl, NH-aryl, NH-heteroaryl, NH-alkyl-cycloalkyl, NH-alkyl-heterocyclyl, NH-alkyl-aryl, NH-alkyl-heteroaryl, NH-alkyl-NH 2 , NH-alkyl-OH, N(alkyl) 2 ,
  • compounds according to the general formula (I) are prepared, wherein the alkyl group is selected from the group consisting of: “methyl, ethyl, n-propyl, 2-propyl, n-butyl, sec.-butyl, tert.-butyl, n-pentyl, iso-pentyl, neo-pentyl, n-hexyl, 2-hexyl, n-octyl, ethylenyl (vinyl), ethynyl, propenyl (—CH2CH ⁇ CH2; —CH ⁇ CH—CH3, —C( ⁇ CH2)-CH3), propinyl (—CH2-C ⁇ CH, —C ⁇ C—CH3), butenyl, butinyl, pentenyl, pentinyl, hexenyl, hexinyl, heptenyl, heptinyl, octeny
  • heterocyclyl group is selected from the group consisting of: “tetrahydrofuryl, tetrahydropyranyl, pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl”.
  • compounds according to the general formula (I) are prepared for the aforementioned use, wherein the heteroaryl group is selected from the group consisting of: “pyrrolyl, furyl, thienyl, thiazolyl, oxazolyl, isoxazolyl, pyrazolyl, imidazolyl, pyridinyl, pyrimidinyl, pyridazinyl, pyrazinyl, phthalazinyl, indolyl, indazolyl, indolizinyl, quinolinyl, isoquinolinyl, quinoxalinyl, quinazolinyl, carbazolyl, phenazinyl, phenothiazinyl, acridinyl”.
  • compounds according to the general formula (I) are prepared for the aforementioned use, wherein the alkyl group is selected from the group consisting of: “methyl, ethyl, n-propyl, 2-propyl, n-butyl, sec.-butyl, tert.-butyl, n-pentyl, iso-pentyl, neo-pentyl, n-hexyl, 2-hexyl, n-octyl, ethylenyl (vinyl), ethynyl, propenyl (—CH2CH ⁇ CH2; —CH ⁇ CH—CH3, —C( ⁇ CH2)-CH3), propinyl (—CH2-C ⁇ CH, —C ⁇ C—CH3), butenyl, butinyl, pentenyl, pentinyl, hexenyl, hexinyl, heptenyl, heptinyl
  • the inventive object was surprisingly achieved in a further aspect by preparing pyridopyrazine compounds selected from the group consisting of:
  • signal transduction pathways selected from the group consisting of: the “ras-Raf-Mek-Erk signal transduction pathway, the PI3K-Akt signal transduction pathway and/or the SAPK signal transduction pathway.
  • alkyl encompasses acyclic saturated or unsaturated hydrocarbon radicals which may be branched or straight-chain and have 1 to 8 carbon atoms, i.e. C 1-8 -alkanyls, C 2-8 -alkenyls and C 2-8 -alkynyls.
  • Alkenyls have at least one C—C double bond and alkynyls at least one C—C triple bond.
  • Alkynyls may additionally also have at least one C—C double bond.
  • Preferred alkyl radicals are methyl, ethyl, n-propyl, 2-propyl, n-butyl, sec-butyl, tert-butyl, n-pentyl, iso-pentyl, neo-pentyl, n-hexyl, 2-hexyl, n-heptyl, n-octyl, n-nonyl, n-decyl, n-undecyl, n-dodecyl, ethylenyl (vinyl), ethynyl, propenyl (—CH 2 CH ⁇ CH 2 ; —CH ⁇ CH—CH 3 , —C( ⁇ CH 2 )—CH 3 ), propynyl (—CH 2 —C ⁇ CH, —C ⁇ C—CH 3 ), butenyl, butynyl, pentenyl, pentynyl, hexeny
  • cycloalkyl means cyclic nonaromatic hydrocarbons having 1 to 3 rings with 3 to 20, preferably 3 to 12 carbon atoms, which may be saturated or unsaturated, more preferably (C 3 -C 8 )cycloalkyl.
  • the cycloalkyl radical may also be part of a bi- or polycyclic system, where, for example, the cycloalkyl radical is fused to an aryl, heteroaryl or heterocyclyl radical as defined herein by any possible and desired ring member(s).
  • the bonding to the compounds of the general formula (I) can be effected via any possible ring member of the cycloalkyl radical.
  • Preferred cycloalkyl radicals are cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclodecyl, cyclohexenyl, cyclopentenyl and cyclooctadienyl.
  • heterocyclyl represents a 3- to 14-membered, preferably 3-, 4-, 5-, 6-, 7- or 8-membered, cyclic organic radical which contains at least 1 heteroatom, optionally 2, 3, 4 or 5 heteroatoms, especially nitrogen, oxygen and/or sulphur, the heteroatoms being the same or different and the cyclic radical being saturated or unsaturated but not aromatic.
  • the heterocyclyl radical may also be part of a bi- or polycyclic system, where, for example, the heterocyclyl radical is fused to an aryl, heteroaryl or cycloalkyl radical as defined herein by any possible and desired ring member(s).
  • heterocyclyl radicals are tetrahydrofuryl, pyrrolidinyl, imidazolidinyl, thiazolidinyl, tetrahydropyranyl, piperidinyl, piperazinyl, morpholinyl, thiapyrrolidinyl, oxapiperazinyl, oxapiperidinyl and oxadiazolyl.
  • aryl means aromatic hydrocarbons having 3 to 14 carbon atoms, preferably 5 to 14 carbon atoms, more preferably 6 to 14 carbon atoms.
  • the aryl radical may also be part of a bi- or polycyclic system, where, for example, the aryl radical is fused to a heterocyclyl, heteroaryl or cycloalkyl radical as defined herein by any possible and desired ring member(s), for example to tetrahydrofuran, tetrahydrothiophene, pyrrolidine, imidazolidine, thiazolidine, tetrahydropyran, dihydropyran, piperidine, furan, thiophene, imidazole, thiazole, oxazole, isoxazole.
  • aryl radicals are phenyl, biphenyl, naphthyl and anthracenyl, but likewise indanyl, indenyl or 1,2,3,4-tetrahydronaphthyl.
  • heteroaryl represents a 5-, 6- or 7-membered cyclic aromatic radical which contains at least 1 heteroatom, if appropriate also 2, 3, 4 or 5 heteroatoms, especially nitrogen, oxygen and/or sulphur, the heteroatoms being the same or different.
  • the number of nitrogen atoms is preferably 0 to 3, that of oxygen and sulphur atoms preferably 0 or 1.
  • the heteroaryl radical may also be part of a bi- or polycyclic system, where, for example, the heteroaryl radical is fused to a heterocyclyl, aryl or cycloalkyl radical as defined herein by any possible and desired ring member(s).
  • heteroaryl radicals are pyrrolyl, furyl, thienyl, thiazolyl, isothiazolyl, oxazolyl, oxadiazolyl, isoxazolyl, pyrazolyl, imidazolyl, triazole, tetrazole, pyridinyl, pyrimidinyl, pyridazinyl, pyrazinyl, triazinyl, phthalazinyl, indolyl, indazolyl, indolizinyl, quinolinyl, isoquinolinyl, quinoxalinyl, quinazolinyl, pteridinyl, carbazolyl, phenazinyl, phenoxazinyl, phenothiazinyl, and acridinyl.
  • alkyl-cycloalkyl mean that alkyl, cycloalkyl, heterocycl, aryl and heteroaryl are each as defined above, and the cycloalkyl, heterocyclyl, aryl and heteroaryl radical is bonded to the compounds of the general formula (I) via an alkyl radical, preferably C 1 -C 8 -alkyl radical, more preferably C 1 -C 4 -alkyl radical.
  • alkyl In connection with “alkyl”, “cycloalkyl”, “heterocyclyl”, “aryl”, “heteroaryl”, alkyl-cycloalkyl”, “alkyl-heterocyclyl”, “alkyl-aryl” and “alkyl-heteroaryl” the term substituted is understood in the sense of this invention unless defined explicitly above in the description and the claims as the substitution of one or more hydrogen groups by F, Cl, Br, I, CN, CF 3 , NH 2 , NH-alkyl, NH-aryl, N(alkyl) 2 , NO 2 , SH, S-alkyl, OH, OCF 3 , O-alkyl, O-aryl, OSO 3 H, OP(O)(OH) 2 , CHO, CO 2 H, SO 3 H or alkyl.
  • the substituents can be the same or different and the substitutions can take place in any arbitrary and possible position of the alkyl, cycloalky
  • halogen encompasses the halogen atoms fluorine, chlorine, bromine and iodine.
  • Multiply substituted groups are to be understood as those which are multiply, e.g. doubly, triply, substituted either at different or at the same atoms, for example, triply substituted at the same C atoms as in the case of CF 3 , —CH 2 CF 3 or at different positions as in the case of —CH(OH)—CH ⁇ CH—CHCl 2 .
  • the multiple substitution can take place with the same or different substituents.
  • the compounds according to the invention have at least one centre of asymmetry, they can be present in the form of their racemates, in the form of the pure enantiomers and/or diastereomers or in the form of mixtures of these enantiomers and/or diastereomers.
  • the mixtures can be present in any arbitrary mixture ratio of the stereoisomers.
  • the compounds according to the invention which have one or a plurality of centres of chirality and which occur as their racemates can be separated into their optical isomers, that is enantiomers or diastereomers, by methods known per se.
  • the separation can be performed by column separation at chiral phases or by recrystallisation from an optically active solvent or by using an optically active acid or base or by derivatisation with an optically active reagent, such as for example, an optically active alcohol and subsequent separation of the residue.
  • inventive compounds may be present in the form of their double bond isomers as “pure” E or Z isomers, or in the form of mixtures of these double bond isomers.
  • the compounds according to the invention can be present in the form of tautomers.
  • the compounds according to the invention can be converted into their physiologically compatible salts using inorganic and organic acids.
  • the pharmaceutically acceptable salts of the compounds according to the invention are preferably formed with hydrochloric acid, bromic acid, sulphuric acid, phosphoric acid, methane sulfonic acid, p-toluene sulfonic acid, carbonic acid, formic acid, acetic acid, trifluoroacetic acid, sulfoacetic acid, oxalic acid, malonic acid, maleic acid, succinic acid, tartaric acid, racemic acid, malic acid, embonic acid, mandelic acid, fumaric acid, lactic acid, citric acid, glutamic acid or asparaginic acid.
  • the salts formed include, among others, hydrochloride, hydrobromide, sulfate, hydrogen sulfate, phosphate, methane sulfonate, tosylate, carbonate, hydrogen carbonate, formiate, acetate, triflate, sulfoacetate, oxalate, malonate, maleate, succinate, tartrate, malate, embonate, mandelate, fumarate, lactate, citrate, glutaminate and aspartate.
  • the stoichiometry of the salts of the compounds according to the invention which are formed can be integer or non-integer multiples of one.
  • the compounds according to the invention can be converted into their physiologically compatible salts using inorganic and organic bases.
  • inorganic bases are, for example, sodium hydroxide, potassium hydroxide, calcium hydroxide
  • possible organic bases are ethanol amine, diethanol amine, triethanol amine, cyclohexylamine, dibenzylethylene diamine and lysine.
  • the stoichiometry of the salts of the compounds according to the invention which are formed can be integer or non-integer multiples of one.
  • solvates and in particular hydrates of the compounds according to the invention which can be obtained, for example, by crystallisation from a solvent or from aqueous solution.
  • solvates and in particular hydrates of the compounds according to the invention can be obtained, for example, by crystallisation from a solvent or from aqueous solution.
  • one, two, three or an arbitrary number of solvate or water molecules can combine with the compounds according to the invention to form solvates and hydrates.
  • the compounds according to the invention can likewise be present in the form of any prodrugs such as, for example, esters, carbonates, carbamates, ureas, amides or phosphates, wherein the actually biologically active form is only released by catabolism.
  • prodrugs such as, for example, esters, carbonates, carbamates, ureas, amides or phosphates, wherein the actually biologically active form is only released by catabolism.
  • the compounds according to the invention can act simultaneously or have a modulating or inhibiting effect on two or more signal transduction pathways or enzymes.
  • the compounds according to the invention can act or have a modulating or inhibiting effect with high selectivity.
  • Such a simultaneous, for example, dual modulation or inhibition of two or more signal transduction pathways e.g. the ras-Raf-Mek-Erk signal pathway, the PI3K-Akt signal pathway and/or the SAPK signal pathway, more especially Erk1/Erk2 and/or PI3K and/or Jnk and/or p38, is advantageous compared with merely single modulation or inhibition of a signal transduction pathway since synergistic therapeutic effects can be brought about, such as for example, intensified apoptosis and faster and more efficient tumour regression.
  • the surprising advantageous effects of the compounds according to the invention allow multiple therapy approaches to be pursued in physiological and/or pathophysiological states or clinical pictures which are sensitive for the treatment or modulation of, or are mediated by, two or more signal transduction pathways.
  • the compounds according to the invention can also act with high selectivity or have a modulating or inhibiting effect on the PI3K-Akt signal transduction pathway or enzymes thereof and that the multiple mechanisms of action and therapy approaches described above can also be used with this signal pathway or enzymes.
  • the compounds according to the invention can also act with high selectivity or have a modulating or inhibiting effect on the SAPK signal transduction pathway or enzymes thereof and that the multiple mechanisms of action and therapy approaches described above can also be used with this signal pathway or enzymes.
  • the compounds according to the invention can also act with high selectivity or have a modulating or inhibiting effect on enzymes such as ATM, ATR, mTOR, DNA-PK and/or hSMG-1 and that the multiple mechanisms of action and therapy approaches described above can also be used with this signal pathway or enzymes.
  • activation is understood according to the invention as follows: “activation, partial activation, inhibition, partial inhibition”. In this case, it is within the specialist knowledge of the average person skilled in the art to measure and determine such activation, partial activation, inhibition, partial inhibition by means of the usual methods of measurement and determination. Thus, a partial activation can be measured and determined in relation to a complete activation; likewise, a partial inhibition in relation to a complete inhibition.
  • inhibiting, inhibition and/or retardation are understood as follows according to the invention: “partial or complete inhibiting, inhibition and/or retardation”. In this case, it is within the specialist knowledge of the average person skilled in the art to measure and determine such inhibiting, inhibition, and/or retardation by means of the usual methods of measurement and determination. Thus, a partial inhibiting, inhibition and/or retardation, for example, can be measured and determined in relation to a complete inhibiting, inhibition and/or retardation.
  • modulation and “inhibiting, inhibition and/or retardation” in connection with “enzymes” and/or “kinases” within the scope of this invention relate both to the inactive form (enzymatically inactive) and/or active form (enzymatically active) of the respective enzyme and/or kinase.
  • the compound according to the invention can have a modulating effect on the inactive form, active form or both forms of the enzyme and/or kinase.
  • the inventive object was surprisingly achieved by preparing the compounds according to the invention which can be used to produce a medicament for the treatment or prevention of physiological and/or pathophysiological states in mammals, wherein the treatment or prevention is effected by modulation of the signal transduction pathway or pathways selected from the group consisting of: the “ras-Raf-Mek-Erk signal transduction pathway, the PI3K-Akt signal transduction pathway and/or the SAPK signal transduction pathway.
  • inventive object was surprisingly achieved by preparing the compounds according to the invention which can be used to produce a medicament for the treatment or prevention of physiological and/or pathophysiological states in mammals, mediated by enzymes selected from the group consisting of: “ATM, ATR, mTOR, DNA-PK, hSMG-1”.
  • the inventive object was surprisingly achieved by preparing the compounds according to the invention which can be used to produce a medicament for the treatment or prevention of physiological and/or pathophysiological states in mammals, wherein the treatment or prevention is effected by modulation of one or more enzymes selected from the group consisting of: “ATM, ATR, mTOR, DNA-PK, hSMG-1”.
  • the compounds according to the invention are prepared for use to produce a medicament for the treatment or prevention of physiological and/or pathophysiological states in mammals mediated by the ras-Raf-Mek-Erk signal transduction pathway, the PI3K-Akt signal transduction pathway and/or use to produce a medicament for the treatment or prevention of physiological and/or pathophysiological states in mammals, wherein the treatment or prevention is effected by modulation of the ras-Raf-Mek-Erk signal transduction pathway and the PI3K-Akt signal transduction pathway.
  • the inventive object was surprisingly achieved by preparing the compounds according to the invention which can be used to produce a medicament for the treatment or prevention of physiological and/or pathophysiological states in mammals, mediated by the PI3K-Akt signal transduction pathway.
  • the inventive object was surprisingly achieved by preparing the compounds according to the invention which can be used to produce a medicament for the treatment or prevention of physiological and/or pathophysiological states in mammals, wherein the treatment or prevention is effected by modulation of the PI3K-Akt signal transduction pathway.
  • the compounds according to the invention are prepared for use to produce a medicament for the treatment and/or prevention of physiological and/or pathophysiological states in mammals mediated by the SAPK signal transduction pathway and the PI3K-Akt signal transduction pathway and/or to produce a medicament for the treatment or prevention of physiological and/or pathophysiological states in mammals, wherein the treatment or prevention is effected by modulation of the SAPK signal transduction pathway and the PI3K-Akt signal transduction pathway.
  • the inventive object was surprisingly achieved by preparing the compounds according to the invention which can be used to produce a medicament for the treatment or prevention of physiological and/or pathophysiological states in mammals, mediated by the SAPK signal transduction pathway.
  • the inventive object was surprisingly achieved by preparing the compounds according to the invention which can be used to produce a medicament for the treatment or prevention of physiological and/or pathophysiological states in mammals, wherein the treatment or prevention is effected by modulation of the SAPK signal transduction pathway.
  • the compounds according to the invention are prepared for the uses described above, wherein the modulation of the ras-Raf-Mek-Erk signal transduction pathway is effected by modulation of one or more enzymes selected from the group consisting of: “tyrosine kinase, serine/threonine kinase, receptor tyrosine kinase, cytoplasmic tyrosine kinase, cytoplasmic serine/threonine kinase” and preferably selected from the group consisting of: “Erk, Erk1, Erk2”.
  • the compounds according to the invention are prepared for the uses described above, wherein the modulation of the P13K-Akt signal transduction pathway is effected by modulation of one or more enzymes selected from the group consisting of: “PI3K, PI3Kalpha, PI3Kbeta, PI3Kgamma, PI3Kdelta, PI3K-C2alpha, PI3K-C2beta, PI3K-Vps34p”.
  • the compounds according to the invention are prepared for the uses described above, wherein the modulation of the SAPK signal transduction pathway is effected by modulation of one or more enzymes selected from the group consisting of: “tyrosine kinase, serine/threonine kinase, receptor tyrosine kinase, cytoplasmic tyrosine kinase, cytoplasmic serine/threonine kinase” and is preferably selected from the group consisting of: “Jnk, Jnk1, Jnk2, Jnk3, p38, p38alpha, p38beta, p38gamma, p38delta”.
  • one or more enzymes selected from the group consisting of: “tyrosine kinase, serine/threonine kinase, receptor tyrosine kinase, cytoplasmic tyrosine kinase, cytoplasmic serine/threonine kin
  • inventive object was surprisingly achieved by preparing the compounds according to the invention according to the aspects, preferred embodiments and uses described above which can be used to produce a medicament for the treatment or prevention of physiological and/or pathophysiological states in mammals, wherein the treatment or prevention is effected by modulation of two or more enzymes.
  • the compounds according to the invention are prepared for the uses described above, wherein in the treatment or prevention effected by modulation of two or more enzymes, at least one enzyme is selected from the group consisting of: “Erk, Erk1, Erk2” and at least one enzyme is selected from the group consisting of: “PI3K, PI3Kalpha, PI3Kbeta, PI3Kgamma, PI3Kdelta, PI3K-C2alpha, PI3K-C2beta, PI3K-Vps34p”.
  • the compounds according to the invention are prepared for the uses described above, wherein in the treatment or prevention effected by modulation of two or more enzymes at least one enzyme is selected from the group consisting of: “Jnk, Jnk1, Jnk2, Jnk3, p38, p38alpha, p38beta, p38gamma, p38delta” and at least one enzyme is selected from the group consisting of: “PI3K, PI3Kalpha, PI3Kbeta, PI3Kgamma, PI3Kdelta, PI3K-C2alpha, PI3K-C2beta, PI3K-Vps34p”.
  • the compounds according to the invention are prepared for the uses described above, wherein in the treatment or prevention effected by modulation of two or more enzymes, at least one enzyme is selected from the group consisting of: “Erk, Erk1, Erk2” and at least one enzyme is selected from the group consisting of: “ATM, ATR, mTOR, DNA-PK, hSMG-1”.
  • the compounds according to the invention are prepared for the uses described above, wherein in the treatment or prevention effected by modulation of two or more enzymes, at least one enzyme is selected from the group consisting of: “Jnk, Jnk1, Jnk2, Jnk3, p38, p38alpha, p38beta, p38gamma, p38delta” and at least one enzyme is selected from the group consisting of: “ATM, ATR, mTOR, DNA-PK, hSMG-1”.
  • the compounds according to the invention are prepared for the uses described above, wherein in the treatment or prevention effected by modulation of two or more enzymes, at least one enzyme is selected from the group consisting of: “PI3K, PI3Kalpha, PI3Kbeta, PI3Kgamma, PI3Kdelta, PI3K-C2alpha, PI3K-C2beta, PI3K-Vps34p” and at least one enzyme is selected from the group consisting of: “ATM, ATR, mTOR, DNA-PK, hSMG-1”.
  • the compounds according to the invention are prepared for the uses described above, wherein the modulation is an inhibition.
  • the compounds according to the invention can be administered within the scope of this invention to all known mammals, in particular, humans, for the treatment and/or prevention.
  • the compounds according to the invention are prepared for the uses described above, wherein the mammal is selected from the group consisting of: “human, domesticated animal, cattle, pet, beef cattle, cow, sheep, pig, goat, horse, pony, donkey, hinny, mule, hare, rabbit, cat, dog, guinea pig, hamster, rat, mouse” and is preferably a human.
  • the compounds according to the invention can be used within the scope of this invention for the treatment and/or prevention of all known physiological and/or pathophysiological states.
  • the compounds according to the invention are prepared for the uses described above, wherein the physiological and/or pathophysiological states are selected from the group consisting of: “malignant tumours, benign tumours, inflammatory diseases, inflammations, pain, rheumatic diseases, arthritic diseases, HIV infections, neurological or neurodegenerative diseases, rheumatism, arthritis, AIDS, ARC (AIDS related complex), Kaposi's sarcoma, tumours originating from the brain and/or nervous system and/or meninges, dementia, Alzheimer's disease, hyperproliferative diseases, psoriasis, endometriosis, scarring, benign prostatahyperplasia (BPH), diseases of the immune system, autoimmune diseases, immunodeficiency diseases, colon tumour, gastric tumour, intestinal tumour, pulmonary tumour, pancreatic tumour, ovarian tumour, prostatic tumour, leukaemia, melanoma, hepatic tumour, renal tumour, head tumour, throat tumour, glioma, breast tumour,
  • the inventive object was surprisingly achieved by preparing the compounds according to the aspects, preferred embodiments and uses described above, for use for the production of a medicament for the treatment or prevention of physiological and/or pathophysiological states in mammals, wherein the medicament comprises at least one further pharmacologically active substance.
  • the inventive object was surprisingly achieved by preparing the compounds according to the aspects, preferred embodiments and uses described above, for use for the production of a medicament for the treatment or prevention of physiological and/or pathophysiological states in mammals, wherein the medicament is administered with at least one further pharmacologically active substance before and/or during and/or after treatment.
  • the inventive object was surprisingly achieved by preparing the compounds according to the aspects, preferred embodiments and uses described above, for use for the production of a medicament for the treatment or prevention of physiological and/or pathophysiological states in mammals, wherein the medicament is administered before and/or during and/or after treatment with radiation therapy and/or surgery.
  • the compounds according to the invention can be administered within the scope of this invention with all known pharmacologically active substances in a combination therapy as described.
  • the compounds according to the invention are prepared for the uses described above, wherein the further pharmacologically active substance is selected from the group consisting of: “DNA topoisomerase I and/or II inhibitors, DNA intercalators, alkylating agents, microtubuli destabilisors, hormone and/or growth factor receptor agonists and/or antagonists, antibodies against growth factors and their receptors, kinase inhibitors, antimetabolites”.
  • the further pharmacologically active substance is selected from the group consisting of: “DNA topoisomerase I and/or II inhibitors, DNA intercalators, alkylating agents, microtubuli destabilisors, hormone and/or growth factor receptor agonists and/or antagonists, antibodies against growth factors and their receptors, kinase inhibitors, antimetabolites”.
  • the compounds according to the invention are prepared for the uses described above, wherein the further pharmacologically active substance is selected from the group consisting of: “asparaginase, bleomycin, carboplatin, carmustin, chlorambucil, cisplatin, colaspase, cyclophosphamide, cytarabine, dacarbazine, dactinomycin, daunorubicin, doxorubicin(adriamycin), epirubicin, etoposide, 5-fluorouracil, hexamethylmelamine, hydroxurea, ifosfamide, irinotecan, leucovorin, lomustine, mechlorethamine, 6-mercaptopurine, mesna, methotrexate, mitomycin C, mitoxantrone, prednisolone, prednisone, procarbazine, raloxifene, streptozocin
  • the inventive object was surprisingly achieved by preparing the pyridopyrazine selected from the group consisting of:
  • Oral administration can take place, for example, in solid form as tablet, capsule, gel capsule, dragee, granule or powder but also in the form of a potable solution.
  • the new compounds according to the invention can be combined with known physiologically compatible adjuvants and excipients usually used, such as gum Arabic, talc, starch, sugar such as, for example, mannite, methyl cellulose, lactose, gelatine, surfactants, magnesium stearate, cyclodextrin, aqueous or non-aqueous excipients, diluents, dispersants, emulsifiers, lubricants, preservatives and flavourings (e.g. ether oils).
  • the compounds according to the invention can also be dispersed in a microparticle, e.g. nanoparticle composition.
  • Non-oral administration can be effected, for example, by intravenous, subcutaneous or intramuscular injection of sterile aqueous or oily solutions, suspensions or emulsions, by means of implants or by ointments, creams or suppositories.
  • administration can be effected as a retard form.
  • Implants can contain inert materials, e.g. biologically degradable polymers or synthetic silicones such as, for example, silicone rubber.
  • Intravaginal administration can be effected by means of vaginal rings, for example.
  • Intrauterine administration can take place, for example, by means of diaphragms or other suitable intrauterine devices.
  • transdermal administration can be provided, in particular by means of a formulation suitable for this purpose and/or suitable means such as plasters, for example.
  • the new compounds according to the invention can also be combined with further pharmaceutically active substances.
  • the individual active constituents can be administered simultaneously or separately and either by the same pathway (e.g. oral) or by separate pathways (e.g. oral and as injection). They can be present or administered in the same or different quantities in a unit dose. A certain dosage regime can be applied insofar as this seems appropriate. In this way, a plurality of the new compounds according to the invention can be combined with one another.
  • the dosage can vary according to the type of indication, the severity of the disease, the type of administration, the age, sex, body weight and sensitivity of the subject to be treated over a wide range. It is within the capabilities of a person skilled in the art to determine a “pharmacologically effective quantity” of the combined pharmaceutical composition.
  • the administration can be made in a single dose or a plurality of separate doses.
  • a suitable unit dose is 0.001 mg to 100 mg of the active substance, i.e. at least one compound according to the invention and optionally a further pharmaceutically active substance, per kg body weight of a patient.
  • compositions comprising a pharmacologically active quantity of at least one compound selected from the group consisting of: “compound 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112, 113, 114, 115, 116, 117, 118, 119, 120,
  • compositions according to the present invention can also contain, in addition to at least one compound according to the invention, as defined previously, at least one further pharmaceutically active substance, as has been described in detail hereinbefore.
  • compositions according to the invention contain at least one of the new compounds according to the invention, as defined hereinbefore, in a pharmacologically active quantity, preferably in a unit dose, e.g. the aforesaid unit dose and preferably in an administration form which allows oral administration.
  • compositions comprising compounds according to the invention and with regard to the use of the compounds according to the invention as medicaments, reference is made to the statements made in connection with the use of the new compounds according to the invention themselves with regard to the possibilities for usage and administration.
  • the inventive object was surprisingly solved by preparing a kit comprising a pharmacologically active quantity of at least one preferred compound according to the invention as presented above and a pharmacologically active quantity of at least one further pharmacologically active substance as defined hereinbefore.
  • Substituted initial stages for selected examples of the pyrido[2,3-b]pyrazine according to the invention can, for example, be obtained according to the procedure in diagram 4 or a corresponding procedure known to the person skilled in the art.
  • Translation of initial stage 16 in diagram 4 into the pyrido[2,3-b]pyrazines substituted according to the invention can, for example, take place according to the procedure in diagram 5 or a corresponding procedure known to the person skilled in the art.
  • the intermediate product 9 in diagram 2 or the intermediate product 11 in diagram 3 can be used or an appropriate substituted intermediate product.
  • cleavage of the corresponding phosphoric ester can be carried out according to procedures known or known to the person skilled in the art using methods known in the literature.
  • the initial compounds and intermediate stages can either be obtained on the market or manufactured according to procedures known to the person skilled in the art.
  • the feed materials 4, 7, 9-16, 21, 24 and 26 represent valuable intermediate compounds for manufacturing pyridopyrazine according to the invention.
  • 2,6-diamino-3-nitropyridine or 2-amino-3,5-dinitro-pyridine is dissolved in a suitable inert solvent, such as methanol, ethanol, dimethylformamide or dioxan, for instance.
  • a suitable inert solvent such as methanol, ethanol, dimethylformamide or dioxan, for instance.
  • the reaction mass is placed in a hydrogen atmosphere with pressure of between 1 and 5 bar being set.
  • the reaction mass is allowed to react for several hours, 1-16 hours, for example, in a temperature range between 20° C. and 60° C.
  • the filter medium can, for example, consist of silica gel, celite or normal glass fibre filters and rewashed using the corresponding solvent.
  • the raw product present in solution is used for the next conversion without additional purification.
  • the 1,2-dione derivative is placed in a suitable, inert solvent, methanol, ethanol, dioxan, toluene or dimethylformamide.
  • a suitable, inert solvent methanol, ethanol, dioxan, toluene or dimethylformamide.
  • 2,3,6-triaminopyridine or 2,3,5-triaminopyridine is added as solution of the raw products in one of the solvents referred to above to the presented 1,2-dione, if necessary adding an acid, such as for example, acetic acid or a base, potassium hydroxide.
  • the reaction mass is allowed to react for a period, between 20 minutes and 40 hours in a temperature range of between 20° C. and 80° C.
  • the filter medium can, for instance, consist of filter paper on the market, rewashed with the corresponding solvent and the remaining solid is dried in vacuum or rather the reaction mass is released from the solvent in vacuum.
  • the reaction mass is stirred into a large quantity of water and the precipitate separated is filtered out or rather the aqueous phase is extracted using a suitable organic solvent, such as, for example, dichloromethane or ethyl acetate and the organic phases concentrated in vacuum.
  • a suitable organic solvent such as, for example, dichloromethane or ethyl acetate and the organic phases concentrated in vacuum.
  • Cleaning of the remaining raw product takes place by recrystallisation from a suitable solvent, dioxan, for example, or using column or rather flash chromatography on silica gel or aluminium oxide.
  • a mixture of methanol and dichloromethane, for example, can serve as solvent.
  • the pyrido-pyrazine on derivate 8 is presented in a suitable, inert solvent, dimethylformamide, dioxan or toluene or without a solvent.
  • a chlorinating agent, phosphoryl chloride or thionyl chloride, for instance, is added at room temperature and the reaction amount is allowed to react in a temperature range of between 20° C. and 100° C. for a period, between 1 hour and 24 hours for example.
  • the reaction mass is poured on to water and neutralised using a suitable aqueous base, caustic soda solution, for instance.
  • the filter medium can consist, for instance of filter paper on the market, is rewashed with the corresponding solvent and the remaining solid dried in vacuum or the aqueous phase is extracted in a suitable organic solvent, such as dichloromethane or ethyl acetate, for example, and the organic phases are concentrated. Cleaning of the remaining raw product takes place by recrystallisation from a suitable solvent, dioxan or toluene, for example, or using column or rather flash chromatography on silica gel or aluminium oxide. A mixture of methanol and dichloromethane, for example, can serve as solvent.
  • 2,3,6-triaminopyridine or 2,3,5-triaminopyridine are presented directly after reduction as a solution of the raw products in one of the solvents referred to above.
  • an oxalic acid derivative such as oxalic acid diethylester or oxalyl chloride
  • the reaction mass is allowed to react, if necessary by adding an acid, such as hydrochloric acid, sulphuric acid or crystalline acid for a period of time, between 10 minutes and 24 hours, for instance, in a temperature range of between 20° C. and 150° C.
  • the filter medium can, for instance, consist of filter paper on the market, rewashed with the corresponding solvent and the remaining solid is dried in vacuum or rather the reaction mass is released from the solvent in vacuum.
  • the reaction mass can be stirred into a large quantity of water and the precipitate separated filtered out or rather the aqueous phase, following neutralisation using an appropriate aqueous base, such as for instance, caustic soda solution, extracted using a suitable organic solvent, such as for instance, dichloromethane or ethyl acetate, and the organic phase concentrated in vacuum.
  • Cleaning of the remaining raw product takes place by recrystallisation from a suitable solvent, dioxan or toluene, for example, or using column or rather flash chromatography on silica gel or aluminium oxide.
  • a suitable solvent dioxan or toluene, for example, or using column or rather flash chromatography on silica gel or aluminium oxide.
  • a mixture of methanol and dichloromethane, for example, can serve as solvent.
  • the diketone derivative 10 is presented in a suitable, inert solvent, dimethylformamide, dioxan or toluene, for instance, or without a solvent.
  • a chlorinating agent, phosphoryl chloride or thionyl chloride, for instance, is added at room temperature and the reaction amount is allowed to react in a temperature range of between 20° C. and 100° C. for a period, between 1 hour and 24 hours, for example.
  • the reaction mass is poured on to water and neutralised using a suitable aqueous base, caustic soda solution, for instance.
  • the filter medium can consist, for instance of filter paper on the market, is rewashed with the corresponding solvent and the remaining solid dried in vacuum or the aqueous phase is extracted in a suitable organic solvent, such as dichloromethane or ethyl acetate, for example, and the organic phases are concentrated in vacuum. Cleaning of the remaining raw product takes place by recrystallisation from a suitable solvent, dioxan or toluene, for example, or using column or flash chromatography on silica gel or aluminium oxide. A mixture of methanol and dichloromethane, for example, can serve as a solvent.
  • the intermediate stage 11 can be converted using a corresponding alcohol, thiol or amine and if necessary using a suitable base, preferably sodium hydride, pyridine, triethylamine, potassium carbonate or sodium methanolate in methanol, in a suitable, inert solvent, such as dimethylformamide, dimethyl sulfoxide, methanol, toluene or in a base as solvent, such as for example, pyridine or triethylamine without solvent.
  • a suitable base preferably sodium hydride, pyridine, triethylamine, potassium carbonate or sodium methanolate in methanol
  • a suitable, inert solvent such as dimethylformamide, dimethyl sulfoxide, methanol, toluene or in a base as solvent, such as for example, pyridine or triethylamine without solvent.
  • the reaction mass is allowed to react for a period, between 30 minutes and 2 days, for example, in a temperature range between 20° C.
  • the intermediate stage 11 can be converted using a corresponding amine and a suitable catalyst, such as for instance, tris(dibenzalacetone) palladium(0) or tetrakis(triphenylphosphine) palladium(0) and a suitable ligand, such as for instance, 2-(dicyclohexylphosphanyl) biphenyl or BINAP, and a suitable base, sodium tert butanolate or potassium carbonate, in a suitable solvent, such as toluene, dioxan or dimethylformamide, for instance.
  • a suitable catalyst such as for instance, tris(dibenzalacetone) palladium(0) or tetrakis(triphenylphosphine) palladium(0) and a suitable ligand, such as for instance, 2-(dicyclohexylphosphanyl) biphenyl or BINAP
  • a suitable base sodium tert butanolate or potassium carbonate
  • a suitable solvent such as tolu
  • the filter medium can, for instance, consist of filter paper on the market, rewashed with the corresponding solvent and the remaining solid is dried in vacuum or the reaction mass is released from the solvent in vacuum.
  • the reaction mass can be stirred into a large quantity of water and the separated precipitate filtered out or the aqueous phase, following neutralisation using a suitable aqueous acid, such as for instance, caustic soda solution, extracted using a suitable organic solvent, such as for example, dichloromethane or ethyl acetate, and the organic phase concentrated in vacuum.
  • Cleaning of the remaining raw product takes place by recrystallisation from a suitable solvent, dioxan or toluene, for example, or using column or flash chromatography on silica gel or aluminium oxide.
  • a suitable solvent dioxan or toluene, for example, or using column or flash chromatography on silica gel or aluminium oxide.
  • a mixture of methanol and dichloromethane, for example, can serve as solvent.
  • the intermediate stage 15 can be converted in the corresponding suitable chloride, bromide, Iodide or tosylate and if necessary using a suitable base, preferably sodium hydride, pyridine, triethylamine, sodium carbonate or sodium methanolate in methanol in a suitable, inert solvent, such as, dimethylformamide, dimethyl sulfoxide, methanol or in a base as solvent, such as, for example, pyridine or triethylamine without a solvent.
  • a suitable base preferably sodium hydride, pyridine, triethylamine, sodium carbonate or sodium methanolate in methanol in a suitable, inert solvent, such as, dimethylformamide, dimethyl sulfoxide, methanol or in a base as solvent, such as, for example, pyridine or triethylamine without a solvent.
  • the reaction mass is allowed to react for a period, between 1 hour and 24 hours for example, in a temperature range between 20° C
  • the intermediate stage 15 can be converted using a corresponding aryl bromide or aryl Iodide and a suitable catalyst, such as, for example, tris(dibenzalacetone)palladium(0) or tetrakis(triphenylphosphine) palladium(0) and a suitable ligand, such as for example, 2-(dicyclohexylphosphanyl) biphenyl or BINAP, and a suitable base, for example, potassium carbonate or sodium tert butanolate or in a suitable solvent, such as toluene, dioxan or dimethylformamide, for instance.
  • a suitable catalyst such as, for example, tris(dibenzalacetone)palladium(0) or tetrakis(triphenylphosphine) palladium(0) and a suitable ligand, such as for example, 2-(dicyclohexylphosphanyl) biphenyl or BINAP
  • a suitable base for example, potassium carbonate
  • the reaction mass is allowed to react for a period, between 10 hours and 30 hours, for example, in a temperature range of between 60° C. and 120° C.
  • the filter medium can, for instance, consist of filter paper on the market, rewashed with the corresponding solvent and the remaining solid is dried in vacuum or the reaction mass is released from the solvent in vacuum.
  • reaction mass can be stirred into a large quantity of water and the precipitate separated filtered out or the aqueous phase, following neutralisation using an suitable aqueous acid, such as for example, caustic soda solution, extracted using a suitable organic solvent, such as, for example, dichloromethane or ethyl acetate, and the organic phase concentrated in vacuum.
  • a suitable aqueous acid such as for example, caustic soda solution
  • organic solvent such as, for example, dichloromethane or ethyl acetate
  • cleaning of the remaining raw product takes place by recrystallisation from a suitable solvent, dioxan or toluene, for example, or using column or flash chromatography on silica gel or aluminium oxide.
  • a mixture of methanol and dichloromethane, for example, can serve as solvent.
  • the products which have come about according to the basic procedure can be translated in consecutive reactions into resultant products according to the invention in a procedure known to the person skilled in the art.
  • the reaction product 16 can be converted using a corresponding isocyanate and if necessary a suitable base, preferably sodium hydride, potassium hexamethyldisalzide, pyridine, triethylamine or potassium carbonate, in a suitable, inert solvent, such as dimethylformamide, dimethyl sulfoxide, acetonitrile, dichloromethane, 1,2-dichloroethane or dioxan or in a base as solvent, such as for example, pyridine or triethylamine without solvent.
  • a suitable base preferably sodium hydride, potassium hexamethyldisalzide, pyridine, triethylamine or potassium carbonate
  • a suitable, inert solvent such as dimethylformamide, dimethyl sulfoxide, acetonitrile, dichloromethane, 1,2-dichloroethane or dioxan or in a base as solvent, such as for example, pyridine or triethylamine without solvent.
  • the filter medium can, for instance, consist of filter paper on the market, rewashed with the appropriate solvent and the remaining solid is dried in vacuum or the reaction mass is released from the solvent in vacuum.
  • the reaction mass can be stirred into a large quantity of water and the precipitate separated filtered out or the aqueous phase, following neutralisation using an appropriate aqueous acid, such as for example, caustic soda solution, extracted using a suitable organic solvent, such as, for instance, dichloromethane or ethyl acetate, and the organic phase concentrated in vacuum.
  • Cleaning of the remaining raw product takes place by recrystallisation from a suitable solvent, dioxan or toluene, for example, or using column or flash chromatography on silica gel or aluminium oxide.
  • a suitable solvent dioxan or toluene, for example, or using column or flash chromatography on silica gel or aluminium oxide.
  • a mixture of methanol and dichloromethane, for example, can serve as a solvent.
  • the reaction product 16 can be converted using phosgene or carbonyldiimidazole and a corresponding amine in a suitable, inert solvent, such as, for example, dimethylformamide, tetrahydrofuran, toluene, dichloromethane or acetonitrile.
  • a suitable base is used, preferably pyridine, sodium hyrdogencarbonate, triethylamine, N-methylmorpholine or sodium acetate.
  • the reaction mass is allowed to react for a period, for example between 15 minutes and 24 hours, in a temperature range between 0 and 60° C.
  • the reaction product 16 can be converted using a corresponding amine phenyl carbamate reagent and if necessary using a suitable base, preferably pyridine, sodium carbonate, triethylamine or sodium hydride, in a suitable, inert solvent, such as, for example, pyridine or triethylamine or without solvent.
  • a suitable base preferably pyridine, sodium carbonate, triethylamine or sodium hydride
  • a suitable, inert solvent such as, for example, pyridine or triethylamine or without solvent.
  • the reaction mass is allowed to react for a period, between 1 hour and 18 hours for example, in a temperature range between 0° C. and 120° C.
  • the filter medium can, for instance, consist of filter paper on the market, rewashed with the corresponding solvent and the remaining solid is dried in vacuum or the reaction mass is released from the solvent in vacuum.
  • reaction mass can be stirred into a large quantity of water and the precipitate separated is filtered out or the aqueous phase, following neutralisation using a suitable aqueous acid, such as for example, caustic soda solution, extracted using a suitable organic solvent, such as, for instance, dichloromethane or ethyl acetate, and the organic phase concentrated in vacuum.
  • a suitable aqueous acid such as for example, caustic soda solution
  • a suitable organic solvent such as, for instance, dichloromethane or ethyl acetate
  • organic phase concentrated in vacuum.
  • Cleaning of the remaining raw product takes place by recrystallisation from a suitable solvent, dioxan or toluene, for example, or using column or flash chromatography on silica gel or aluminium oxide.
  • a mixture of methanol and dichloromethane, for example, can serve as solvent.
  • the reaction product 16 can be translated using a corresponding isothiocyanate, and if necessary, a suitable base, preferably sodium hydride, triethylamine, potassium carbonate or pyridine, in a suitable, inert solvent, such as for instance dimethylformamide, tetrahydrofuran, acetone or toluene, or in a base as solvent, such as, for instance, pyridine or triethylamine or without solvent.
  • a suitable base preferably sodium hydride, triethylamine, potassium carbonate or pyridine
  • a suitable, inert solvent such as for instance dimethylformamide, tetrahydrofuran, acetone or toluene
  • a base as solvent such as, for instance, pyridine or triethylamine or without solvent.
  • the reaction mass is allowed to react for a period, for example between 30 minutes and 90 hours, in a temperature range between 0 and 115° C.,
  • the filter medium can, for instance, consist of filter paper on the market, rewashed with the corresponding solvent and the remaining solid is dried in vacuum or the reaction mass is released from the solvent in vacuum.
  • reaction mass can be stirred into a large quantity of water and the precipitate separated filtered out or the aqueous phase, following neutralisation using a suitable aqueous acid, such as for example, caustic soda solution, extracted using a suitable organic solvent, such as, for instance, dichloromethane or ethyl acetate, and the organic phase concentrated in vacuum.
  • a suitable aqueous acid such as for example, caustic soda solution
  • organic solvent such as, for instance, dichloromethane or ethyl acetate
  • cleaning of the remaining raw product takes place by recrystallisation from a suitable solvent, dioxan or toluene, for example, or using column or flash chromatography on silica gel or aluminium oxide.
  • a mixture of methanol and dichloromethane, for example, can serve as a solvent.
  • the reaction product 16 can be translated using thiophosgene or thiocarbonyl diimidazole and a corresponding amine in a suitable, inert solvent, such as dimethylformamide, tetrahydrofuran, toluene, dichloromethane, ethanol or acetonitrile.
  • a suitable base preferably pyridine, sodium hydrogencarbonate, potassium carbonate, triethylamine or imidazole is used.
  • the reaction mixture is allowed to react for several hours, between 1 and 24 hours for example, in a temperature range between ⁇ 10 and 80° C.
  • the filter medium can, for instance, consist of filter paper on the market, rewashed with the appropriate solvent and the remaining solid is dried in vacuum or the reaction mass is released from the solvent in vacuum.
  • the reaction mass can be stirred into a large quantity of water and the precipitate separated filtered out or the aqueous phase, following neutralisation using an appropriate aqueous acid, such as for example, caustic soda solution, extracted using a suitable organic solvent, such as, for instance, dichloromethane or ethyl acetate, and the organic phase concentrated in vacuum.
  • Cleaning of the remaining raw product takes place by recrystallisation from a suitable solvent, dioxan or toluene, for example, or using column or flash chromatography on silica gel or aluminium oxide.
  • a suitable solvent dioxan or toluene, for example, or using column or flash chromatography on silica gel or aluminium oxide.
  • a mixture of methanol and dichloromethane, for example, can serve as solvent.
  • the products which have come about according to the basic procedure can be translated in consecutive reactions into resultant products according to the invention in a procedure known to the person skilled in the art.
  • the reaction product 21 can be translated using the corresponding aryl/heteroaryl boronic acid derivatives or aryl/heteroaryl organotin compounds and a suitable catalyst, such as, for example, Pd(PPh 3 ) 4 , [1,1′-bis(diphenylphosphino)ferrocene]dichloro palladium(II) or Pd 2 (dba) 3 and a suitable base, for example, sodium carbonate, cesium carbonate or triethylamine, in a suitable solvent, such as for instance, dimethylformamide, dimethylformamide/water, toluene, acetonitrile, dimethoxyethane or dioxan.
  • a suitable catalyst such as, for example, Pd(PPh 3 ) 4 , [1,1′-bis(diphenylphosphino)ferrocene]dichloro palladium(II) or Pd 2 (dba) 3
  • a suitable base for example,
  • the reaction mixture is allowed to react for a period, for instance between 6 hours and several days, in a temperature range between 60° C. and 120° C.
  • the filter medium can, for instance, consist of filter paper on the market, rewashed with the corresponding solvent and the remaining solid is dried in vacuum or the reaction mass is released from the solvent in vacuum.
  • reaction mass can be stirred into a large quantity of water and the precipitate separated filtered out or the aqueous phase, following neutralisation using a suitable aqueous acid, such as for example, caustic soda solution, extracted using a suitable organic solvent, such as, for instance, dichloromethane or ethyl acetate, and the organic phase concentrated in vacuum.
  • a suitable aqueous acid such as for example, caustic soda solution
  • organic solvent such as, for instance, dichloromethane or ethyl acetate
  • cleaning of the remaining raw product takes place by recrystallisation from a suitable solvent, dioxan or toluene, for example, or using column or flash chromatography on silica gel or aluminium oxide.
  • a mixture of methanol and dichloromethane, for example, can serve as a solvent.
  • the reaction product 21 can be converted using corresponding alkyl zinc halogenides and a suitable catalyst, such as, for example, Pd(PPh 3 ) 4 , [1,1′-bis(diphenylphosphino)ferrocene]dichloro palladium(II) or PdCl 2 (PPh 3 ) 2 in a suitable solvent, such as for instance, dimethylformamide, tetrahydrofuran, toluene, dimethoxyethane or dioxan.
  • a suitable solvent such as for instance, dimethylformamide, tetrahydrofuran, toluene, dimethoxyethane or dioxan.
  • the reaction mixture is allowed to react for a period, for example between 30 minutes and 48 hours, in a temperature range between room temperature and 120° C.
  • the filter medium can, for instance, consist of filter paper on the market, rewashed with the appropriate solvent and the remaining solid is dried in vacuum or the reaction mass is released from the solvent in vacuum.
  • the reaction mass can be stirred into a large quantity of water and the perpetrate separated is filtered out or the aqueous phase, following neutralisation using an appropriate aqueous acid, such as for example, dichloromethane or ethyl acetate, and the organic phase concentrated in vacuum.
  • Cleaning of the remaining raw product takes place by recrystallisation from a suitable solvent, dioxan or toluene, for example, or using column or flash chromatography on silica gel or aluminium oxide.
  • a mixture of methanol and dichloromethane, for example, can serve as solvent.
  • the products which have come about according to the basic procedure can be translated in consecutive reactions into resultant products according to the invention in a procedure known to the person skilled in the art.
  • the reaction product 24 can be converted, for example, using a corresponding chloride, bromide, Iodide and if necessary using a suitable base, preferably sodium hydride, pyridine, triethylamine, sodium carbonate or sodium methanolate in methanol in a suitable, inert solvent, such as, dimethylformamide, dimethyl sulfoxide, methanol, dioxan, tetrahydrofuran, toluene or in a base as solvent, such as, for instance, pyridine or triethylamine or without solvent.
  • a suitable base preferably sodium hydride, pyridine, triethylamine, sodium carbonate or sodium methanolate in methanol in a suitable, inert solvent, such as, dimethylformamide, dimethyl sulfoxide, methanol, dioxan, tetrahydrofuran, toluene or in a base as solvent, such as, for instance, pyridine or triethyl
  • reaction mass is allowed to react for a period, between 30 minutes and 2 days, for example, in a temperature range between 0° C. and 140° C.
  • an amino substituted intermediate stage 24 can be used, using a corresponding chloride, bromide or Iodide and a suitable catalyst, such as, for instance, tris(dibenzalacetone)palladium(0) or tetrakis(triphenylphosphine) palladium(0) and a suitable ligand, such as, for instance, 2-(dicyclohexylphosphanyl) biphenyl or BINAP, and a suitable base, sodium tert butanolate or potassium carbonate, in a suitable solvent, such as toluene, dioxan or dimethylformamide, for instance.
  • a suitable catalyst such as, for instance, tris(dibenzalacetone)palladium(0) or tetrakis(triphenylphosphine) palladium(0) and a suitable ligand, such as
  • the reaction mass is allowed to react for a period, between 2 hours and 30 hours, for example, in a temperature range of between 60° C. and 120° C.
  • the filter medium can, for instance, consist of filter paper on the market, rewashed with the suitable solvent and the remaining solid is dried in vacuum or the reaction mass is released from the solvent in vacuum.
  • reaction mass can be stirred into a large quantity of water and the perpetrate separated filtered out or the aqueous phase, following neutralisation using a suitable aqueous acid, such as for instance, caustic soda solution, extracted using a suitable organic solvent, such as, for example, dichloromethane or ethyl acetate, and the organic phase concentrated in vacuum.
  • a suitable aqueous acid such as for instance, caustic soda solution
  • organic solvent such as, for example, dichloromethane or ethyl acetate
  • cleaning of the remaining raw product takes place by recrystallisation from a suitable solvent, dioxan or toluene, for example, or using column or flash chromatography on silica gel or aluminium oxide.
  • a mixture of methanol and dichloromethane, for example, can serve as solvent.
  • the reaction product 26 can be converted, for example, using a corresponding chloro-phosphoric ester and if necessary using a suitable base, preferably sodium hydride, pyridine, triethylamine, potassium carbonate or lithium diisopropylamide in a suitable, inert solvent, such as for example, dimethylformamide, dimethyl sulfoxide, methanol, dioxan, tetrahydrofuran, toluene or in a base as solvent, such as for example, pyridine or triethylamine or without solvent.
  • a suitable base preferably sodium hydride, pyridine, triethylamine, potassium carbonate or lithium diisopropylamide
  • a suitable, inert solvent such as for example, dimethylformamide, dimethyl sulfoxide, methanol, dioxan, tetrahydrofuran, toluene or in a base as solvent, such as for example, pyridine or triethylamine or without solvent
  • the reaction mass is allowed to react for a period, between 1 hour and 24 hours for example, in a temperature range between 0° C. and 100° C.
  • the filter medium can, for instance, consist of filter paper on the market, rewashed with the suitable solvent and the remaining solid is dried in vacuum or the reaction mass is released from the solvent in vacuum.
  • the reaction mass can be stirred into a large quantity of water and the precipitate separated filtered out or the aqueous phase, following neutralisation using a suitable aqueous acid, such as for example, caustic soda solution, extracted using a suitable organic solvent, such as, for example, dichloromethane or ethyl acetate, and the organic phase concentrated in vacuum.
  • Cleaning of the remaining raw product takes place by recrystallisation from a suitable solvent, dioxan or toluene, for example, or using column or rather flash chromatography on silica gel or aluminium oxide.
  • a suitable solvent dioxan or toluene, for example, or using column or rather flash chromatography on silica gel or aluminium oxide.
  • a mixture of methanol and dichloromethane, for example, can serve as solvent.
  • OH, SH and NH 2 groups can possibly undergo unwanted secondary reactions. It is therefore preferable to provide them with protective groups or in the case of NH 2 to replace with NO 2 and subsequently to split the protective group off or to reduce the NO 2 group.
  • at least one OH group for example, can be replaced by a benzyl oxy group and/or at least one SH group, for example, by an S-benzyl group and/or at least one NH 2 group by a NH-benzyl group or by an NO 2 group.
  • At least one—preferably all—benzyl oxy group(s) or NH benzyl group(s) can be split off, for instance, using hydrogen and palladium on carbon and/or at least one—preferably all—S-benzyl group(s), for example, using sodium in ammonia and/or at least one—preferably all NO 2 group(s) are reduced, for instance using hydrogen and Raney nickel to NH 2
  • the initial compounds and intermediate stages with at least one COOH group can be converted into carbonic ester or carboxamide derivatives by transformation using an activator, such as thionyl chloride or carbonyldiimidazole and a subsequent transformation using a suitable alcohol or amine.
  • an activator such as thionyl chloride or carbonyldiimidazole
  • a suitable alcohol or amine e.g. benzyl alcohol
  • at least one—preferably all—carbonic ester or carboxamide group(s) in the initial compounds and intermediate stages can be split, for instance using diluted aqueous acids or bases to free at least one—preferably all —OH group(s) and/or NH 2 group(s) and/or COOH groups.
  • Conversion using amines can be undertaken using triethylamine as a base in dioxan or without a base in dioxan. Additional compounds according to the invention were produced according to this method: For example, the compounds 59, 62, 65, 81 and 88.
  • the inhibitory effect of the compounds according to the invention was tested on various serine/threonine, tyrosine and lipid kinases in enzymatic assays.
  • Recombinant human kinases such as, for example, Erk2, PI3Kalpha, -beta, -gamma, -delta, p38alpha, p38gamma, Jnk1, Jnk2 and others were used in this case, partly as full-length kinases, partly as shortened fragments, but at least consisting of the functional kinase domains.
  • the commercial kinase proteins (Proqinase, Upstate) were used as recombinant fusion proteins with GST (glutathion-S-transferase) or His-Tag. Depending on the type of substrate, the various kinase reactions were quantified by means of suitable ALPHATM beads (Perkin-Elmer).
  • MAPK-ALPHAs e.g. Erk2
  • Erk2 the test substance
  • 10 ⁇ M ATP and 15 nM biotinylated MBP (myelin basic protein) substrate were incubated on a 384-well Optiplate (Perkin-Elmer) in a volume of 15 ⁇ l for 1 h in 25 mM Tris, 10 mM MgCl 2 , 0.1% Tween-20, 100 ⁇ M NaVO 4 , 2 mM DTT at pH 7.5.
  • kinase reaction was then stopped by adding 10 ⁇ l of the ALPHA bead mixes (10 ⁇ g/ml, #6760617/Perkin-Elmer) pre-incubated with anti-phospho MBP antibody (320 pM, #05-429/Upstate) in 25 mM Tris, 200 mM NaCl, 110 nM EDTA and 0.3% BSA and left to stand overnight.
  • PI3K-ALPHAs e.g.
  • PI3Kalpha the test substance, 1 ng PI3Kalpha (#14-602, Upstate), 100 ⁇ M ATP and 20 ⁇ M PIP 2 substrate (#P4508, Echelon) were incubated on a 384-well Optiplate (Perkin-Elmer) for 1 h in 50 mM Hepes, 50 mM NaCl, 5 mM MgCl 2 , 0.05% Chaps, 5 mM DTT at pH 7.4.
  • the kinase reaction was then stopped by adding ALPHA bead mixes (10 ⁇ g/ml, #6760603/Perkin-Elmer) pre-incubated with 1 nM GST:Grp1 fusion protein (Upstate) and 15 nM biotinylated PIP3 (#C-39B6/Echelon) in 50 mM Hepes, 50 mM NaCl, 50 mM EDTA and 0.1% BSA and left to stand overnight.
  • ALPHA bead mixes (10 ⁇ g/ml, #6760603/Perkin-Elmer) pre-incubated with 1 nM GST:Grp1 fusion protein (Upstate) and 15 nM biotinylated PIP3 (#C-39B6/Echelon) in 50 mM Hepes, 50 mM NaCl, 50 mM EDTA and 0.1% BSA and left to stand overnight.
  • the fluorescence was detected the following morning in a FusionTM a ⁇ -system (Perkin-Elmer).
  • the %-inhibition values per substance concentration were calculated by means of the following formula from the raw data determined in the FusionTM :
  • the compounds according to the invention showed effective inhibition of Erk, PI3K, p38alpha and Jnk1+Jnk2 with IC 50 values of up to 6 nM (see Table 2).
  • the principle of this test is based on the intracellular reduction of the tetrazolium dye XTT (sodium 3′-[1-(phenylaminocarbonyl)-3,4-tetrazolium]-bis(4-methoxy-6-nitro)benzene sulfonic acid, Sigma) to a formazan dye by mitochondrial dehydrogenases.
  • the dye is only formed by metabolically active cells and its photometrically measurable intensity is a quantitative indicator for the presence of living cells.
  • the reduction of dye formation by incubation of the cells with substances serves as a parameter for the anti-proliferative effect.
  • tumour cell lines were injected into 96-well microtitre plates in a defined cell number (5000 cells/well for BxPC3 and Hct116; 10000 cells/well for MDA MB468) and then incubated overnight in an incubator at 37° C., 5% CO 2 and 95% air humidity.
  • the test substances were prepared as stock solutions (10 mM) in DMSO.
  • the potential inhibitor substances were added to the cells in quarter-logarithmically graded dilutions, resulting in final concentrations of 0.28 ⁇ M-50 ⁇ M.
  • the cell plates were then incubated for 45 h in an incubator at 37° C., 5% CO 2 and 95% air humidity.
  • the substrate XTT was mixed with PMS (N-Methyl dibenzopyrazine methylsulfate, Sigma) and added to the cells so that a final concentration of 325 ⁇ g XTT/ml and 2.5 ⁇ g PMS/ml was obtained. It was then incubated for 3 h at 37° C., 95% air humidity. The formazan salt formed by the cellular dehydrogenases could then be quantified by adsorption at 490 nm.
  • PMS N-Methyl dibenzopyrazine methylsulfate
  • the % inhibition value was evaluated by means of the following formula from the values for the optical densities measured in each case at 490 nm:
  • the compounds according to the invention showed partly effective inhibition of the cell proliferation with EC 50 values of to ⁇ 1 ⁇ M (see Table 3).
  • This method can be used to predict whether the kinase modulator under study also achieves the desired effect in a cellular context, i.e. in this case, a substrate protein downstream of the target kinase is investigated for its phosphorylation status.
  • the cells incubated with substance are lysed and the total protein separated on a reducing polyacrylamide gel.
  • the proteins are then transferred to a PVDF membrane by means of Western Blotting and the substrate bands sought are made visible with specific antibodies and a suitable detection method.
  • the substrate proteins downstream of the target kinases are detected simultaneously with a respectively special anti-phospho-antibody and at the same time a total antibody which recognizes the substrate total protein.
  • This simultaneous measurement can be made using the duplex technology of the ODYSSEY imagers (LiCOR).
  • the intensity of the total substrate bands is used for normalising or quantifying the phosphorylation inhibition or activation.
  • Suitable tumour cell lines (e.g. BxPC3, Hct116 or MDA MB468) were injected into six-well microtitre plates in a defined cell number (e.g. 350 000 cells/well for BxPC3 and Hct116) in the respective standard complete medium and then incubated overnight at 37° C., 5% CO 2 and 95% air humidity. The cells were then incubated for a further 24 h under serum-reduced conditions i.e. in the respective medium but with only 0.25% serum. The test substances were prepared as stock solutions (10 mM) in DMSO and incubated with the cells in final concentrations of 5, 15.8 and 50 ⁇ M for 5 h.
  • a defined cell number e.g. 350 000 cells/well for BxPC3 and Hct116
  • the test substances were prepared as stock solutions (10 mM) in DMSO and incubated with the cells in final concentrations of 5, 15.8 and 50 ⁇ M for 5 h.
  • the compounds according to the invention exhibited dual inhibition of Erk (MAPK1/2) and PI3K (see Table 4) which are indicated by inhibition of the band intensity of both corresponding phospho-substrate proteins Rsk1 and Akt.
  • the reduction of the fluorescence intensity of the phospho-substrate bands (pRsk and pAkt) is given in the table below as % inhibition and relates to the following formula:
  • the band intensity (fluorescence intensity) of the respective non-inhibited (without substance) phospho-substrates was used as 100% control.
  • the PI3K inhibitor Ly294002 known from the literature showed only weak PI3K inhibition compared to the pyridopyrazine derivatives, i.e. inhibition of the PI3K substrate p-Akt and as predicted, no Erk inhibition or inhibition of the Erk substrate p-Rsk.
  • Wortmannin a further PI3K inhibitor known from the literature—showed complete inhibition of the PI3K substrate pAkt, but no Erk or p-RSK inhibition.
  • both reference substances used here showed no dual inhibition, i.e. of Erk and PI3K simultaneously but only a PI3K inhibition.

Abstract

Pyridopyrazine compounds which are suitable for the treatment or prevention of physiological and/or pathophysiological states mediated and/or modulated by signal transduction pathways and/or enzymes in mammals and in particular in humans. A compound according to the general formula (I)
Figure US08937068-20150120-C00001

Description

CROSS-REFERENCE TO RELATED APPLICATION
This application claims priority to U.S. 60/735,707, filed Nov. 11, 2005, which is hereby incorporated by reference in its entirety.
TECHNICAL FIELD
The present invention relates to pyridopyrazine derivatives with new biological action and their use for the treatment of physiological and/or pathophysiological states mediated and/or modulated by signal transduction pathways in mammals and in particular in humans.
PRIOR ART
The signal transduction cascades ras-Raf-Mek-Erk and PI3K-Akt play a central role in cell growth, cell proliferation, apoptosis, adhesion, migration and glucose metabolism. Consequently, the fundamental involvement in the pathogenesis of diseases such as cancer, neurodegeneration and inflammatory diseases is proven both for the ras-Raf-Mek-Erk and for the PI3K-Akt signal pathway. The individual components of these signal cascades are therefore important therapeutic points of attack for intervention in various disease processes (Weinstein-Oppenheimer C. R. et al. 2000, Chang F. et al. 2003, Katso R. et al 2001 and Lu Y. et al 2003).
The molecular and biochemical properties of both signal pathways are first described separately hereinafter.
A plurality of growth factors, cytokines and oncogenes transduce their growth-promoting signals via the activation of G-protein coupled ras which leads to the activation of serine threonine kinase Raf and to the activation of mitogen-activated protein kinase kinase 1 and 2 (MAPKK1/2 or Mek1/2) and results in the phosphorylation and activation of MAPK 1 and 2—also known as extracellular signal regulated kinase (Erk1 and 2). Compared to other signal pathways, the ras-Raf-Mek-Erk signal pathway combines a large number of proto-oncogenes, including ligands, tyrosine kinase receptors, G-proteins, kinases and nuclear transcription factors. Tyrosine kinases such as, for example, EGFR (Mendelsohn J. et al., 2000) frequently mediate constitutively active signals to the downstream ras-Raf-Mek-Erk signal pathway in tumour events caused by overexpression and mutation. Ras mutations are mutated in 30% of all human tumours (Khleif S. N. et al., 1999, Marshall C., 1999), the highest incidence of 90% being found in pancreatic carcinomas (Friess H. et al., 1996, Sirivatanauksorn V. et al., 1998). For c-Raf a deregulated expression and/or activation has been described in various tumours (Hoshino R. et al., 1999, McPhillips F. et al., 2001). B-Raf point mutants were detected in 66% of all human malignant melanomas, 14% of all ovarian carcinomas and 12% of all carcinomas of the colon (Davies H. et al., 2002). It is therefore not surprising that Erk1/2 is primarily involved in many cellular processes such as cell growth, cell proliferation and cell differentiation (Lewis T. S. et al., 1998, Chang F. et al., 2003).
In addition, the members of the Raf kinases also have Mek-Erk-indepedent anti-apoptotic functions whose molecular steps have not yet been fully described. Ask1, Bcl-2, Akt and Bag1 have been described as possible interaction partners for the Mek-Erk-independent Raf activity (Chen J et al., 2001, Troppmaier J. et al., 2003, Rapp U. R. et al., 2004, Gotz R. et al., 2005). It is assumed nowadays that both Mek-Erk-dependent and Mek-Erk-independent signal transduction mechanisms control the activation of the upstream ras and Raf stimuli.
The isoenzymes of the phosphatidylinositol 3-kinases (PI3Ks) function predominantly as lipid kinases and catalyse the D3 phosphorylation of the second-messenger lipids PtdIns (phosphatidylinositol) to PtdIns(3)P, PtdIns(3,4)P2, PtdIns(3,4,5)P3 phosphatidylinositol phosphates. The class I PI3Ks are composed structurally of the catalytic (p110alpha, beta, gamma, delta) and the regulatory (p85alpha, beta or p101gamma) subunits. Furthermore, the class II (PI3K-C2alpha, PI3K-C2beta) and class III (Vps34p) enzymes belong to the family of the P13 kinases (Wymann M. P. et al., 1998, VanHaesebroeck B. et al., 2001). The PIP increase triggered by the PI3Ks activates the proliferative ras-Raf-Mek-Erk signal pathway via the coupling of ras on the one hand (Rodriguez-Viciana P. et al., 1994) and on the other hand stimulates the anti-apoptotic signal pathway by recruiting Akt to the cell membrane and consequent overactivation of this kinase (Alessi D. R. et al., 1996, Chang H. W. et al., 1997, Moore S. M. et al., 1998). Consequently, the activation of PI3Ks fulfils at least two crucial mechanisms for tumour formation, namely the activation of cell growth and cell differentiation and the inhibition of apoptosis. In addition, PI3K also have protein-phosphorylating properties (Dhand et al., 1994, Bondeva T. et al., 1998, Bondev A. et al., 1999, VanHaesebroeck B. et al., 1999) which can trigger a PI3Ks-intrinsically regulating serine autophosphorylation for example. In addition, it is known that PI3Ks also have kinase-independent regulating effector properties, e.g. during control of cardiac contraction (Crackower M. A. et al., 2002, Patrucco et al., 2004). It is furthermore proven that PI3Kdelta and PI3Kgamma are specifically expressed on hematopoietic cells and therefore constitute potential points of attack for isoenzyme-specific PI3Kdelta and PI3Kgamma inhibitors in the treatment of inflammatory diseases such as rheumatism, asthma and allergies and in the treatment of B and T cell lymphomas (Okkenhaug K. et al., 2003, Ali K. et al., 2004, Sujobert P. et al., 2005). PI3Kalpha, which was recently identified as a proto-oncogene (Shayesteh L. et al., 1999, Ma Y. Y. et al., 2000, Samuels Y. et al., 2004, Campbell I. G. et al., 2004, Levine D. A., 2005) is considered to be an important target in the treatment of tumour diseases. The importance of PI3K species as a target for the development of active substances is therefore extremely diverse (Chang F. & Lee J. T. et al, 2003).
The kinases related to PI3K (PIKK), which include the serine/threonine kinases mTOR, ATM, ATR, h-SMG-1 and DNA-PK (Chiang G. G. et al 2004) are also of great interest. Their catalytic domains have a high sequence homology to the catalytic domains of PI3Ks.
In addition, the loss of the tumour suppressor protein PTEN (Li J. et al., 1997, Steck P. A. et al., 1997)—whose function is the reversion of the phosphorylation initiated by the PI3K—contributes to an overactivation of Akt and its downstream cascade components and thereby emphasise the causal importance of PI3K as a target molecule for tumour therapy.
Various inhibitors of individual components of the ras-Raf-Mek-Erk and PI3K-Akt signal pathways have already been published and patented.
The present state of development in the field of kinase inhibitors, in particular of the ras-Raf-Mek-Erk and PI3K-Akt pathway, is described in the reviews of J. S. Sebolt-Leopold et al., 2004, and R. Wetzker et al., 2004. These publications contain comprehensive listings of the published patent specifications which describe the synthesis and use of low-molecular ras-Raf-Mek-Erk- and PI3K inhibitors.
The kinase inhibitor Bay 43-9006 (WO 99/32111, WO 03/068223) which is already undergoing clinical trials, shows a relatively non-specific inhibition pattern of serine/threonine and of tyrosine kinases such as Raf, VEGFR2/3, Flt-3, PDGFR, c-Kit and other kinases. Great importance is attached to this inhibitor in angiogenesis-induced advanced tumour diseases (e.g. in renal cell carcinoma) and also in melanomas having a high B-Raf mutation rate. The clinical effect of Bay 43-9006 is currently also being determined in patients presenting with refractory solid tumours in combination with, for example, Docetaxel. Mild side effects and promising anti-tumoral effects have been described so far. No inhibition of the kinases in the PI3K-Akt signal pathway has been described for Bay 43-9006.
The Mek1/2 inhibitor PD0325901 (WO 02/06213) is currently undergoing a Phase I clinical trial. The precursor substance CI-1040 (WO 00/35435, WO 00/37141) was striking because of its high Mek specificity and target affinity. However, this compound proved to be metabolically unstable in Phase I/II trials. No clinical data are available yet for the current successor substance PD0325901. However, no interaction with Erk1 or Erk2 nor any PI3K-Akt signal pathway inhibiting function or its simultaneous modulation has yet been disclosed for this Mek inhibitor.
Patent specifications WO 04/104002 and WO 04/104003 describe pyrido[2,3-b]pyrazines, which can be substituted in the 6- or 7-position with urea, thiourea, amidine or guanidine groups. These compounds possess properties as inhibitors or modulators of kinases, in particular of tyrosine and serine/threonine kinases, and a use as a medicament is specified. However, no use of these compounds as modulators of lipid kinases, alone or in combination with tyrosine and serine/threonine kinases has been described.
In addition, patent specification WO 99/17759 describes pyrido[2,3-b]pyrazines which, among other things, carry alkyl-, aryl- and heteroaryl-substituted carbamates in the 6-position. These compounds are to be used to modulate serine threonine protein kinases.
Patent specification WO 05/007099 describes, among other things, urea-substituted pyrido[2,3-b]pyrazines as inhibitors of the serine/threonine kinase PKB. A use in the treatment of cancer diseases is specified for these compounds. However, no specific examples of urea-substituted pyridopyrazines with these biological properties are given.
Further examples of pyrido[2,3-b]pyrazines substituted with urea in the 6- and 7-position are given in patent specification WO 05/056547. The compounds in this patent specification are described as inhibitors of protein kinases, in particular GSK-3, Syk and JAK-3. A use in the treatment of proliferative diseases is given for these compounds among other things. No use of these compounds as modulators of lipid kinases, alone or in combination with serine/threonine kinases is described.
The patent application WO 04/005472 describes, among other things pyrido[2,3-b]pyrazines substituted with carbamate in the 6-position which inhibit the growth of bacteria as antibacterial substances. No antitumour effect is described.
Certain diphenyl quinoxalines and pyrido[2,3-b]pyrazines with special alkylpyrrolidine, alkylpiperidine or alkyl sulfonamides group at a phenyl ring which can additionally also bear urea or carbamate substitutions in the 6- or 7-position are described in patent specifications WO 03/084473, WO 03/086394 and WO 03/086403 as inhibitors of the activity of the serine/threonine kinase Akt. A use in the treatment of cancer diseases is specified for these compounds. No defined indication of a biological effect is given for the pyrido[2,3-b]pyrazine compounds described therein as examples.
Patent specification WO 03/024448 describes amide and acrylamide-substituted pyrido[2,3-b]pyrazines which can also contain carbamates as additional substituents and can be used as histone deacetylase inhibitors for the treatment of cell proliferation diseases.
In another publication (Temple C. et al. 1990) the synthesis of a 6-ethylcarbamate-substituted pyrido[2,3-b]pyrazine derivative is described as one example. No antitumour effect is disclosed or made obvious.
The synthesis of further derivatives of 6-ethylcarbamate-substituted pyrido[2,3-b]pyrazine is described in a publication by R. D. Elliott (J. Org. Chem. 1968). No biological effect of these compounds is described or disclosed.
The publication by C. Temple (1968) describes the synthesis and investigation of 6-ethylcarbamate-substituted pyrido[2,3-b]pyrazines as potential antimalarial drugs. No antitumour effect is disclosed or made obvious.
The PI3K inhibitors published so far are undergoing preclinical trials. ICOS disclosed a PI3K inhibitor IC87114 with high PI3Kdelta isoenzyme specificity (WO 01/81346). For PI103 (WO 04/017950) Yamanouchi/Piramed describe a selectivity versus the PI3Kalpha isoform. In addition, a research environment which has attracted a great deal of attention exists in the early development of PI3K inhibitors (see the review of R. Wetzker et al., 2004).
Inhibitors of the SAPK signal pathway, either of Jnk or of p38 have been described in the literature (Gum R. J., 1998, Bennett B. L. et al 2001, Davies S. P. et al 2000). However, no PI3K-inhibiting function and also no specific inhibition of Erk1 or Erk2 or simultaneous inhibition of SAPKs, Erk1, Erk2, or PI3Ks is disclosed for these SAPK inhibitors.
DESCRIPTION OF THE INVENTION
The object of the present invention is to provide new compounds which can be used for the treatment or prevention of physiological and/or pathophysiological states in mammals, in particular in humans, which are mediated by signal transduction pathways selected from the group consisting of: “ras-Raf-Mek-Erk signal transduction pathway, PI3K-Akt signal transduction pathway and/or SAPK signal transduction pathway”. A further object of the invention is to provide new compounds for the aforesaid uses by modulation of said signal transduction pathways. It is further the object of the present invention to provide new compounds which can be used for the treatment or prevention of physiological and/or pathophysiological states in mammals, in particular in humans, which are mediated by enzymes selected from the group consisting of: “ATM, ATR, mTOR, DNA-PK, hSMG-1”. A further object of the invention is to provide new compounds for the aforesaid uses by modulation of said enzymes.
The inventive object was surprisingly achieved in one aspect by preparing a compound according to the general formula (I)
Figure US08937068-20150120-C00002

wherein the substituents R1, R2, R3, R4 have the following meaning:
R1 and R can be independently of one another
(i) hydrogen
(ii) hydroxyl
(iii) halogen
(iv) alkyl, wherein the alkyl group is saturated and can consists of 1 to 8 C atoms,
(v) unsubstituted or substituted aryl, wherein the alkyl group can be substituted with one or more, the same or different F, Cl, Br, I, CF3, CN, NH2, NH-alkyl, NH-cycloalkyl, NH-heterocyclyl, NH-aryl, NH-heteroaryl, NH-alkyl-cycloalkyl, NH-alkyl-heterocyclyl, NH-alkyl-aryl, NH-alkyl-heteroaryl, NH-alkyl-NH2, NH-alkyl-OH, N(alkyl)2, NHC(O)-alkyl, NHC(O)-cycloalkyl, NHC(O)-heterocyclyl, NHC(O)-aryl, NHC(O)-heteroaryl, NHC(O)-alkyl-aryl, NHC(O)-alkyl-heteroaryl, NHSO2-alkyl, NHSO2-cycloalkyl, NHSO2-heterocyclyl, NHSO2-aryl, NHSO2-heteroaryl, NHSO2-alkyl-aryl, NHSO2-alkyl-heteroaryl, NO2, SH, S-alkyl, S-aryl, S-heteroaryl, OH, OCF3, O-alkyl, O-cycloalkyl, O-heterocyclyl, O-aryl, O-heteroaryl, O-alkyl-cycloalkyl, O-alkyl-heterocyclyl, O-alkyl-aryl, O-alkyl-heteroaryl, O-alkyl-OH, O—(CH2)n—O, OC(O)-alkyl, OC(O)-cycloalkyl, OC(O)-heterocyclyl, OC(O)-aryl, OC(O)-heteroaryl, OC(O)-alkyl-aryl, OC(O)-alkyl-heteroaryl, OSO3H, OSO2-alkyl, OSO2-cycloalkyl, OSO2-heterocyclyl, OSO2-aryl, OSO2-heteroaryl, OSO2-alkyl-aryl, OSO2-alkyl-heteroaryl, OP(O)(OH)2, C(O)-alkyl, C(O)-aryl, C(O)-heteroaryl, CO2H, CO2-alkyl, CO2-cycloalkyl, CO2-heterocyclyl, CO2-aryl, CO2-heteroaryl, CO2-alkyl-cycloalkyl, CO2-alkyl-heterocyclyl, CO2-alkyl-aryl, CO2-alkyl-heteroaryl, C(O)—NH2, C(O)NH-alkyl, C(O)NH-cycloalkyl, C(O)NH-heterocyclyl, C(O)NH-aryl, C(O)NH-heteroaryl, C(O)NH-alkyl-cycloalkyl, C(O)NH-alkyl-heterocyclyl, C(O)NH-alkyl-aryl, C(O)NH-alkyl-heteroaryl, C(O)N(alkyl)2, C(O)N(cycloalkyl)2, C(O)N(aryl)2, C(O)N(heteroaryl)2, SO-alkyl, SO-aryl, SO2-alkyl, SO2-aryl, SO2NH2, SO2NH-alkyl, SO2NH-aryl, SO2NH-heteroaryl, SO2NH-alkyl-aryl, SO3H, SO2O-alkyl, SO2O-aryl, SO2O-alkyl-aryl, alkyl, cycloalkyl, heterocyclyl, aryl or heteroaryl, n can have the value 1, 2 or 3 and the alkyl-, cycloalkyl-, heterocyclyl-, aryl-, heteroaryl-, alkyl-cycloalkyl-, alkyl-heterocyclyl-, alkyl-aryl- and alkyl-heteroaryl substituents for their part can in turn be substituted,
(vi) unsubstituted or substituted heteroaryl, wherein the heteroaryl group can be substituted with one or more, the same or different F, Cl, Br, I, CF3, CN, NH2, NH-alkyl, NH-cycloalkyl, NH-heterocyclyl, NH-aryl, NH-heteroaryl, NH-alkyl-cycloalkyl, NH-alkyl-heterocyclyl, NH-alkyl-aryl, NH-alkyl-heteroaryl, NH-alkyl-NH2, NH-alkyl-OH, N(alkyl)2, NHC(O)-alkyl, NHC(O)-cycloalkyl, NHC(O)-heterocyclyl, NHC(O)-aryl, NHC(O)-heteroaryl, NHC(O)-alkyl-aryl, NHC(O)-alkyl-heteroaryl, NHSO2-alkyl, NHSO2-cycloalkyl, NHSO2-heterocyclyl, NHSO2-aryl, NHSO2-heteroaryl, NHSO2-alkyl-aryl, NHSO2-alkyl-heteroaryl, NO2, SH, S-alkyl, S-aryl, S-heteroaryl, OH, OCF3, O-alkyl, O-cycloalkyl, O-aryl, O-heteroaryl, O-alkyl-cycloalkyl, O-alkyl-heterocyclyl, O-alkyl-aryl, O-alkyl-heteroaryl, OC(O)-alkyl, OC(O)-cycloalkyl, OC(O)-heterocyclyl, OC(O)-aryl, OC(O)-heteroaryl, OC(O)-alkyl-aryl, OC(O)-alkyl-heteroaryl, OSO3H, OSO2-alkyl, OSO2-cycloalkyl, OSO2-heterocyclyl, OSO2-aryl, OSO2-heteroaryl, OSO2-alkyl-aryl, OSO2-alkyl-heteroaryl, OP(O)(OH)2, C(O)-alkyl, C(O)-aryl, C(O)-heteroaryl, CO2H, CO2-alkyl, CO2-cycloalkyl, CO2-heterocyclyl, CO2-aryl, CO2-heteroaryl, CO2-alkyl-cycloalkyl, CO2-alkyl-heterocyclyl, CO2-alkyl-aryl, CO2-alkyl-heteroaryl, C(O)—NH2, C(O)NH-alkyl, C(O)NH-cycloalkyl, C(O)NH-heterocyclyl, C(O)NH-aryl, C(O)NH-heteroaryl, C(O)NH-alkyl-cycloalkyl, C(O)NH-alkyl-heterocyclyl, C(O)NH-alkyl-aryl, C(O)NH-alkyl-heteroaryl, C(O)N(alkyl)2, C(O)N(cycloalkyl)2, C(O)N(aryl)2, C(O)N(heteroaryl)2, SO2NH2, SO2NH-alkyl, SO2NH-aryl, SO2NH-heteroaryl, SO2NH-alkyl-aryl, SO3H, SO2O-alkyl, SO2O-aryl, SO2O-alkyl-aryl, alkyl, cycloalkyl, heterocyclyl, aryl or heteroaryl, and the alkyl-, cycloalkyl-, heterocyclyl-, aryl- and heteroaryl substituents for their part can in turn be substituted,
(vii) OR5, wherein R5 can be alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, alkyl-cycloalkyl, alkyl-heterocyclyl, alkyl-aryl or alkyl-heteroaryl, and the alkyl-, cycloalkyl-, heterocyclyl-, aryl-, heteroaryl-, alkyl-cycloalkyl, alkyl-heterocyclyl, alkyl-aryl or alkyl-heteroaryl substituents for their part can in turn be substituted,
(viii) SR6, wherein R6 can be alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, alkyl-cycloalkyl, alkyl-heterocyclyl, alkyl-aryl or alkyl-heteroaryl and the alkyl-, cycloalkyl-, heterocyclyl-, aryl- and heteroaryl-, alkyl-cycloalkyl-, alkyl-heterocyclyl-, alkyl-aryl- or alkyl-heteroaryl substituents for their part can in turn be substituted,
(ix) NR7R8, wherein R7 and R8 independently of one another can be hydrogen, alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, alkyl-cycloalkyl, alkyl-heterocyclyl, alkyl-aryl or alkyl-heteroaryl and the alkyl-, cycloalkyl-, heterocyclyl-, aryl- and heteroaryl-, alkyl-cycloalkyl, alkyl-heterocyclyl, alkyl-aryl or alkyl-heteroaryl substituents for their part can in turn be substituted,
or R7 and R8 together mean cycloalkyl or heterocyclyl, wherein cycloalkyl and heterocyclyl for their part can in turn be substituted.
R3 and R4 can independently of one another mean hydrogen or NR9R10 assuming that if R3=NR9R110, R4=H, and if R4=NR9R110, R3=H,
wherein R9 can be hydrogen, alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, alkyl-cycloalkyl, alkyl-heterocyclyl, alkyl-aryl or alkyl-heteroaryl and the alkyl-, cycloalkyl-, heterocyclyl-, aryl- and heteroaryl-, alkyl-cycloalkyl, alkyl-heterocyclyl, alkyl-aryl or alkyl-heteroaryl substituents for their part can in turn be substituted,
and R10:
can mean —C(Y)NR11R12, wherein Y═O, S and R11 and R12 can be independently of one another
(i) hydrogen,
(ii) unsubstituted or substituted alkyl wherein the alkyl group can be substituted with one or more, the same or different F, Cl, Br, I, CF3, CN, NH2, NH-alkyl, NH-cycloalkyl, NH-heterocyclyl, NH-aryl, NH-heteroaryl, NH-alkyl-cycloalkyl, NH-alkyl-heterocyclyl, NH-alkyl-aryl, NH-alkyl-heteroaryl, N(alkyl)2, NHC(O)-alkyl, NHC(O)-cycloalkyl, NHC(O)-heterocyclyl, NHC(O)-aryl, NHC(O)-heteroaryl, NHC(O)-alkyl-aryl, NHC(O)-alkyl-heteroaryl, NHSO2-alkyl, NHSO2-cycloalkyl, NHSO2-heterocyclyl, NHSO2-aryl, NHSO2-heteroaryl, NHSO2-alkyl-aryl, NHSO2-alkyl-heteroaryl, NO2, SH, S-alkyl, S-cycloalkyl, S-heterocyclyl, S-aryl, S-heteroaryl, OH, OCF3, O-alkyl, O-cycloalkyl, O-heterocyclyl, O-aryl, O-heteroaryl, O-alkyl-cycloalkyl, O-alkyl-heterocyclyl, O-alkyl-aryl, O-alkyl-heteroaryl, OC(O)-alkyl, OC(O)-cycloalkyl, OC(O)-heterocyclyl, OC(O)-aryl, OC(O)-heteroaryl, OC(O)-alkyl-aryl, OC(O)-alkyl-heteroaryl, OSO3H, OSO2-alkyl, OSO2-cycloalkyl, OSO2-heterocyclyl, OSO2-aryl, OSO2-heteroaryl, OSO2-alkyl-aryl, OSO2-alkyl-heteroaryl, OP(O)(OH)2, C(O)-alkyl, C(O)-aryl, C(O)-heteroaryl, CO2H, CO2-alkyl, CO2-cycloalkyl, CO2-heterocyclyl, CO2-aryl, CO2-heteroaryl, CO2-alkyl-cycloalkyl, CO2-alkyl-heterocyclyl, CO2-alkyl-aryl, CO2-alkyl-heteroaryl, C(O)—NH2, C(O)NH-alkyl, C(O)NH-cycloalkyl, C(O)NH-heterocyclyl, C(O)NH-aryl, C(O)NH-heteroaryl, C(O)NH-alkyl-cycloalkyl, C(O)NH-alkyl-heterocyclyl, C(O)NH-alkyl-aryl, C(O)NH-alkyl-heteroaryl, C(O)N(alkyl)2, C(O)N(cycloalkyl)2, C(O)N(aryl)2, C(O)N(heteroaryl)2, SO-alkyl, SO-aryl, SO2-alkyl, SO2-aryl, SO2NH2, SO2NH-alkyl, SO2NH-aryl, SO2NH-heteroaryl, SO2NH-alkyl-aryl, SO3H, SO2O-alkyl, SO2O-aryl, SO2O-alkyl-aryl, cycloalkyl, heterocyclyl, aryl or heteroaryl,
(iii) unsubstituted or substituted cycloalkyl, wherein the cycloalkyl group can be substituted with one or more, the same or different F, Cl, Br, I, NH2, NH-alkyl, NH-cycloalkyl, NH-heterocyclyl, NH-aryl, NH-heteroaryl, NH-alkyl-aryl, NH-alkyl-heteroaryl, N(alkyl)2, NHC(O)-alkyl, NHC(O)-cycloalkyl, NHC(O)-heterocyclyl, NHC(O)-aryl, NHC(O)-heteroaryl, NHC(O)-alkyl-aryl, NHC(O)-alkyl-heteroaryl, NHSO2-alkyl, NHSO2-cycloalkyl, NHSO2-heterocyclyl, NHSO2-aryl, NHSO2-heteroaryl, NHSO2-alkyl-aryl, NHSO2-alkyl-heteroaryl, OH, O-alkyl, O-cycloalkyl, O-heterocyclyl, O-aryl, O-heteroaryl, O-alkyl-aryl, O-alkyl-heteroaryl, OC(O)-alkyl, OC(O)-cycloalkyl, OC(O)-heterocyclyl, OC(O)-aryl, OC(O)-heteroaryl, OC(O)-alkyl-aryl, OC(O)-alkyl-heteroaryl, OSO3H, OSO2-alkyl, OSO2-cycloalkyl, OSO2-heterocyclyl, OSO2-aryl, OSO2-heteroaryl, OSO2-alkyl-aryl, OSO2-alkyl-heteroaryl, OP(O)(OH)2, CO2H, CO2-alkyl, CO2-cycloalkyl, CO2-heterocyclyl, CO2-aryl, CO2-heteroaryl, CO2-alkyl-cycloalkyl, CO2-alkyl-heterocyclyl, CO2-alkyl-aryl, CO2-alkyl-heteroaryl, C(O)—NH2, C(O)NH-alkyl, C(O)NH-cycloalkyl, C(O)NH-heterocyclyl, C(O)NH-aryl, C(O)NH-heteroaryl, C(O)NH-alkyl-cycloalkyl, C(O)NH-alkyl-heterocyclyl, C(O)NH-alkyl-aryl, C(O)NH-alkyl-heteroaryl, C(O)N(alkyl)2, C(O)N(cycloalkyl)2, C(O)N(aryl)2, C(O)N(heteroaryl)2, alkyl, or aryl,
(iv) unsubstituted or substituted heterocyclyl, wherein the heterocyclyl group can be substituted with one or more, the same or different OH, O-alkyl, O-aryl, NH2, NH-alkyl, NH-aryl, alkyl, alkyl-aryl or aryl,
(v) unsubstituted or substituted aryl, wherein the aryl group can be substituted with one or more, the same or different F, Cl, Br, I, CF3, CN, NH2, NH-alkyl, NH-cycloalkyl, NH-heterocyclyl, NH-aryl, NH-heteroaryl, NH-alkyl-cycloalkyl, NH-alkyl-heterocyclyl, NH-alkyl-aryl, NH-alkyl-heteroaryl, NH-alkyl-NH2, NH-alkyl-OH, N(alkyl)2, NHC(O)-alkyl, NHC(O)-cycloalkyl, NHC(O)-heterocyclyl, NHC(O)-aryl, NHC(O)-heteroaryl, NHC(O)-alkyl-aryl, NHC(O)-alkyl-heteroaryl, NHSO2-alkyl, NHSO2-cycloalkyl, NHSO2-heterocyclyl, NHSO2-aryl, NHSO2-heteroaryl, NHSO2-alkyl-aryl, NHSO2-alkyl-heteroaryl, NO2, SH, S-alkyl, S-cycloalkyl, S-heterocyclyl, S-aryl, S-heteroaryl, OH, OCF3, O-alkyl, O-cycloalkyl, O-heterocyclyl, O-aryl, O-heteroaryl, O-alkyl-cycloalkyl, O-alkyl-heterocyclyl, O-alkyl-aryl, O-alkyl-heteroaryl, O-alkyl-OH, O—(CH2)n—O, OC(O)-alkyl, OC(O)-cycloalkyl, OC(O)-heterocyclyl, OC(O)-aryl, OC(O)-heteroaryl, OC(O)-alkyl-aryl, OC(O)-alkyl-heteroaryl, OSO3H, OSO2-alkyl, OSO2-cycloalkyl, OSO2-heterocyclyl, OSO2-aryl, OSO2-heteroaryl, OSO2-alkyl-aryl, OSO2-alkyl-heteroaryl, OP(O)(OH)2, C(O)-alkyl, C(O)-aryl, C(O)-heteroaryl, CO2H, CO2-alkyl, CO2-cycloalkyl, CO2-heterocyclyl, CO2-aryl, CO2-heteroaryl, CO2-alkyl-cycloalkyl, CO2-alkyl-heterocyclyl, CO2-alkyl-aryl, CO2-alkyl-heteroaryl, C(O)—NH2, C(O)NH-alkyl, C(O)NH-cycloalkyl, C(O)NH-heterocyclyl, C(O)NH-aryl, C(O)NH-heteroaryl, C(O)NH-alkyl-cycloalkyl, C(O)NH-alkyl-heterocyclyl, C(O)NH-alkyl-aryl, C(O)NH-alkyl-heteroaryl, C(O)N(alkyl)2, C(O)N(cycloalkyl)2, C(O)N(aryl)2, C(O)N(heteroaryl)2, SO-alkyl, SO-aryl, SO2-alkyl, SO2-aryl, SO2NH2, SO2NH-alkyl, SO2NH-aryl, SO2NH-heteroaryl, SO2NH-alkyl-aryl, SO3H, SO2O-alkyl, SO2O-aryl, SO2O-alkyl-aryl, alkyl, cycloalkyl, heterocyclyl, aryl or heteroaryl, and n can have the value 1, 2 or 3,
(vi) unsubstituted or substituted heteroaryl, wherein the heteroaryl group can be substituted with one or more, the same or different F, Cl, Br, I, CF3, CN, NH2, NH-alkyl, NH-cycloalkyl, NH-heterocyclyl, NH-aryl, NH-heteroaryl, NH-alkyl-cycloalkyl, NH-alkyl-heterocyclyl, NH-alkyl-aryl, NH-alkyl-heteroaryl, NH-alkyl-NH2, NH-alkyl-OH, N(alkyl)2, NHC(O)-alkyl, NHC(O)-cycloalkyl, NHC(O)-heterocyclyl, NHC(O)-aryl, NHC(O)-heteroaryl, NHC(O)-alkyl-aryl, NHC(O)-alkyl-heteroaryl, NHSO2-alkyl, NHSO2-cycloalkyl, NHSO2-heterocyclyl, NHSO2-aryl, NHSO2-heteroaryl, NHSO2-alkyl-aryl, NHSO2-alkyl-heteroaryl, NO2, SH, S-alkyl, S-aryl, S-heteroaryl, OH, OCF3, O-alkyl, O-cycloalkyl, O-aryl, O-heteroaryl, O-alkyl-cycloalkyl, O-alkyl-heterocyclyl, O-alkyl-aryl, O-alkyl-heteroaryl, OC(O)-alkyl, OC(O)-cycloalkyl, OC(O)-heterocyclyl, OC(O)-aryl, OC(O)-heteroaryl, OC(O)-alkyl-aryl, OC(O)-alkyl-heteroaryl, OSO3H, OSO2-alkyl, OSO2-cycloalkyl, OSO2-heterocyclyl, OSO2-aryl, OSO2-heteroaryl, OSO2-alkyl-aryl, OSO2-alkyl-heteroaryl, OP(O)(OH)2, C(O)-alkyl, C(O)-aryl, C(O)-heteroaryl, CO2H, CO2-alkyl, CO2-cycloalkyl, CO2-heterocyclyl, CO2-aryl, CO2-heteroaryl, CO2-alkyl-cycloalkyl, CO2-alkyl-heterocyclyl, CO2-alkyl-aryl, CO2-alkyl-heteroaryl, C(O)—NH2, C(O)NH-alkyl, C(O)NH-cycloalkyl, C(O)NH-heterocyclyl, C(O)NH-aryl, C(O)NH-heteroaryl, C(O)NH-alkyl-cycloalkyl, C(O)NH-alkyl-heterocyclyl, C(O)NH-alkyl-aryl, C(O)NH-alkyl-heteroaryl, C(O)N(alkyl)2, C(O)N(cycloalkyl)2, C(O)N(aryl)2, C(O)N(heteroaryl)2, SO2NH2, SO2NH-alkyl, SO2NH-aryl, SO2NH-heteroaryl, SO2NH-alkyl-aryl, SO3H, SO2O-alkyl, SO2O-aryl, SO2O-alkyl-aryl, alkyl, cycloalkyl, heterocyclyl, aryl or heteroaryl,
(vii) —C(O)—R17, wherein R17 can be alkyl, aryl or heteroaryl and the alkyl and aryl substituents for their part can in turn be substituted,
(viii) or R11 and R12 together can mean cycloalkyl or heterocyclyl,
can mean —C(Y)NR13R14 wherein Y═NH and R13 and R14 can be independently of one another,
(i) hydrogen,
(ii) unsubstituted or substituted aryl, wherein the aryl group can be substituted with one or more, the same or different F, Cl, Br, I, CF3, CN, NH2, NH-alkyl, NH-cycloalkyl, NH-heterocyclyl, NH-aryl, NH-heteroaryl, NH-alkyl-aryl, NH-alkyl-heteroaryl, N(alkyl)2, NHC(O)-alkyl, NHC(O)-cycloalkyl, NHC(O)-heterocyclyl, NHC(O)-aryl, NHC(O)-heteroaryl, NHSO2-alkyl, NHSO2-cycloalkyl, NHSO2-aryl, NHSO2-heteroaryl, NO2, SH, S-alkyl, S-cycloalkyl, S-heterocyclyl, S-aryl, S-heteroaryl, OH, OCF3, O-alkyl, O-cycloalkyl, O-heterocyclyl, O-aryl, O-heteroaryl, O-alkyl-cycloalkyl, O-alkyl-aryl, O-alkyl-heteroaryl, OC(O)-alkyl, OC(O)-cycloalkyl, OC(O)-heterocyclyl, OC(O)-aryl, OC(O)-heteroaryl, OSO2-alkyl, OSO2-cycloalkyl, OSO2-aryl, OSO2-heteroaryl, C(O)-alkyl, C(O)-aryl, CO2H, CO2-alkyl, CO2-cycloalkyl, CO2-heterocyclyl, CO2-aryl, CO2-heteroaryl, CO2-alkyl-cycloalkyl, CO2-alkyl-heterocyclyl, CO2-alkyl-aryl, CO2-alkyl-heteroaryl, C(O)—NH2, C(O)NH-alkyl, C(O)NH-cycloalkyl, C(O)NH-heterocyclyl, C(O)NH-aryl, C(O)NH-heteroaryl, C(O)NH-alkyl-cycloalkyl, C(O)NH-alkyl-heterocyclyl, C(O)NH-alkyl-aryl, C(O)NH-alkyl-heteroaryl, C(O)N(alkyl)2, C(O)N(cycloalkyl)2, C(O)N(aryl)2, C(O)N(heteroaryl)2, SO-alkyl, SO-aryl, SO2-alkyl, SO2-aryl, SO2NH2, SO3H, alkyl, cycloalkyl, heterocyclyl, aryl or heteroaryl,
(iii) unsubstituted or substituted cycloalkyl, wherein the cycloalkyl group can be substituted with one or more, the same or different F, Cl, Br, I, NH2, NH-alkyl, NH-cycloalkyl, NH-heterocyclyl, NH-aryl, NH-heteroaryl, NH-alkyl-aryl, NH-alkyl-heteroaryl, N(alkyl)2, NHC(O)-alkyl, NHC(O)-cycloalkyl, NHC(O)-heterocyclyl, NHC(O)-aryl, NHC(O)-heteroaryl, NHSO2-alkyl, NHSO2-cycloalkyl, NHSO2-aryl, NHSO2-heteroaryl, OH, O-alkyl, O-cycloalkyl, O-heterocyclyl, O-aryl, O-heteroaryl, O-alkyl-aryl, O-alkyl-heteroaryl, OC(O)-alkyl, OC(O)-cycloalkyl, OC(O)-heterocyclyl, OC(O)-aryl, OC(O)-heteroaryl, OSO2-alkyl, OSO2-cycloalkyl, OSO2-aryl, OSO2-heteroaryl, CO2H, CO2-alkyl, CO2-cycloalkyl, CO2-heterocyclyl, CO2-aryl, CO2-heteroaryl, C(O)—NH2, C(O)NH-alkyl, C(O)NH-cycloalkyl, C(O)NH-heterocyclyl, C(O)NH-aryl, C(O)NH-heteroaryl, C(O)NH-alkyl-aryl, C(O)NH-alkyl-heteroaryl, C(O)N(alkyl)2, alkyl, or aryl,
(iv) unsubstituted or substituted heterocyclyl, wherein the heterocyclyl group can be substituted with one or more, the same or different OH, O-alkyl, O-aryl, NH2, NH-alkyl, NH-aryl, alkyl, or aryl,
(v) unsubstituted or substituted aryl, wherein the aryl group can be substituted with one or more, the same or different F, Cl, Br, I, CF3, CN, NH2, NH-alkyl, NH-cycloalkyl, NH-heterocyclyl, NH-aryl, NH-heteroaryl, NH-alkyl-cycloalkyl, NH-alkyl-heterocyclyl, NH-alkyl-aryl, NH-alkyl-heteroaryl, NH-alkyl-NH2, NH-alkyl-OH, N(alkyl)2, NHC(O)-alkyl, NHC(O)-cycloalkyl, NHC(O)-heterocyclyl, NHC(O)-aryl, NHC(O)-heteroaryl, NHSO2-alkyl, NHSO2-aryl, NHSO2-heteroaryl, NO2, SH, S-alkyl, S-cycloalkyl, S-heterocyclyl, S-aryl, S-heteroaryl, OH, OCF3, O-alkyl, O-cycloalkyl, O-heterocyclyl, O-aryl, O-heteroaryl, O-alkyl-cycloalkyl, O-alkyl-heterocyclyl, O-alkyl-aryl, O-alkyl-heteroaryl, O-alkyl-OH, O—(CH2)n—O, OC(O)-alkyl, OC(O)-cycloalkyl, OC(O)-heterocyclyl, OC(O)-aryl, OC(O)-heteroaryl, OSO2-alkyl, OSO2-cycloalkyl, OSO2-aryl, OSO2-heteroaryl, C(O)-alkyl, C(O)-aryl, C(O)-heteroaryl, CO2H, CO2-alkyl, CO2-cycloalkyl, CO2-heterocyclyl, CO2-aryl, CO2-heteroaryl, CO2-alkyl-cycloalkyl, CO2-alkyl-heterocyclyl, CO2-alkyl-aryl, CO2-alkyl-heteroaryl, C(O)—NH2, C(O)NH-alkyl, C(O)NH-cycloalkyl, C(O)NH-heterocyclyl, C(O)NH-aryl, C(O)NH-heteroaryl, C(O)NH-alkyl-cycloalkyl, C(O)NH-alkyl-heterocyclyl, C(O)NH-alkyl-aryl, C(O)NH-alkyl-heteroaryl, C(O)N(alkyl)2, C(O)N(cycloalkyl)2, C(O)N(aryl)2, C(O)N(heteroaryl)2, SO-alkyl, SO-aryl, SO2-alkyl, SO2-aryl, SO2NH2, SO2NH-alkyl, SO2NH-aryl, SO2NH-heteroaryl, SO3H, SO2O-alkyl, SO2O-aryl, SO2O-heteroaryl, alkyl, cycloalkyl, heterocyclyl, aryl or heteroaryl, and n can have the value 1, 2 or 3,
(vi) unsubstituted or substituted heteroaryl, wherein the heteroaryl group can be substituted with one or more, the same or different F, Cl, Br, I, CF3, CN, NH2, NH-alkyl, NH-cycloalkyl, NH-heterocyclyl, NH-aryl, NH-heteroaryl, NH-alkyl-aryl, NH-alkyl-heteroaryl, N(alkyl)2, NHC(O)-alkyl, NHC(O)-cycloalkyl, NHC(O)-heterocyclyl, NHC(O)-aryl, NHC(O)-heteroaryl, NHSO2-alkyl, NHSO2-aryl, NHSO2-heteroaryl, NO2, SH, S-alkyl, S-aryl, OH, OCF3, O-alkyl, O-cycloalkyl, O-heterocyclyl, O-aryl, O-heteroaryl, OC(O)-alkyl, OC(O)-cycloalkyl, OC(O)-heterocyclyl, OC(O)-aryl, OC(O)-heteroaryl, OSO2-alkyl, OSO2-cycloalkyl, OSO2-aryl, OSO2-heteroaryl, C(O)-alkyl, C(O)-aryl, C(O)-heteroaryl, CO2H, CO2-alkyl, CO2-cycloalkyl, CO2-heterocyclyl, CO2-aryl, CO2-heteroaryl, CO2-alkyl-cycloalkyl, CO2-alkyl-heterocyclyl, CO2-alkyl-aryl, CO2-alkyl-heteroaryl, C(O)—NH2, C(O)NH-alkyl, C(O)NH-cycloalkyl, C(O)NH-heterocyclyl, C(O)NH-aryl, C(O)NH-heteroaryl, C(O)NH-alkyl-cycloalkyl, C(O)NH-alkyl-heterocyclyl, C(O)NH-alkyl-aryl, C(O)NH-alkyl-heteroaryl, C(O)N(alkyl)2, C(O)N(cycloalkyl)2, C(O)N(aryl)2, C(O)N(heteroaryl)2, SO2-alkyl, SO2-aryl, SO2NH2, SO2NH-alkyl, SO2NH-aryl, SO2NH-heteroaryl, SO3H, SO2O-alkyl, SO2O-aryl, SO2O-heteroaryl, alkyl, cycloalkyl, heterocyclyl, aryl or heteroaryl,
(vii) or R13 and R14 together can mean cycloalkyl or heterocyclyl,
can mean —C(NR15)R16, wherein R15=H and R16 can be
(i) unsubstituted or substituted alkyl, wherein the alkyl group can be substituted with one or more, the same or different F, Cl, Br, I, CF3, NH2, NH-alkyl, NH-cycloalkyl, NH-heterocyclyl, NH-aryl, NH-heteroaryl, NH-alkyl-aryl, NH-alkyl-heteroaryl, N(alkyl)2, NHC(O)-alkyl, NHC(O)-cycloalkyl, NHC(O)-heterocyclyl, NHC(O)-aryl, NHC(O)-heteroaryl, NHSO2-alkyl, NHSO2-cycloalkyl, NHSO2-aryl, NHSO2-heteroaryl, NO2, SH, S-alkyl, S-cycloalkyl, S-heterocyclyl, S-aryl, S-heteroaryl, OH, OCF3, O-alkyl, O-cycloalkyl, O-heterocyclyl, O-aryl, O-heteroaryl, O-alkyl-cycloalkyl, O-alkyl-aryl, O-alkyl-heteroaryl, OC(O)-alkyl, OC(O)-cycloalkyl, OC(O)-heterocyclyl, OC(O)-aryl, OC(O)-heteroaryl, OSO2-alkyl, OSO2-cycloalkyl, OSO2-aryl, OSO2-heteroaryl, C(O)-alkyl, C(O)-aryl, CO2H, CO2-alkyl, CO2-cycloalkyl, CO2-heterocyclyl, CO2-aryl, CO2-heteroaryl, CO2-alkyl-cycloalkyl, CO2-alkyl-heterocyclyl, CO2-alkyl-aryl, CO2-alkyl-heteroaryl, C(O)—NH2, C(O)NH-alkyl, C(O)NH-cycloalkyl, C(O)NH-heterocyclyl, C(O)NH-aryl, C(O)NH-heteroaryl, C(O)NH-alkyl-cycloalkyl, C(O)NH-alkyl-heterocyclyl, C(O)NH-alkyl-aryl, C(O)NH-alkyl-heteroaryl, C(O)N(alkyl)2, C(O)N(cycloalkyl)2, C(O)N(aryl)2, C(O)N(heteroaryl)2, SO-alkyl, SO-aryl, SO2-alkyl, SO2-aryl, SO2NH2, SO3H, alkyl, cycloalkyl, heterocyclyl, aryl or heteroaryl,
(ii) unsubstituted or substituted cycloalkyl, wherein the cycloalkyl group can be substituted with one or more, the same or different F, Cl, Br, I, NH2, NH-alkyl, NH-cycloalkyl, NH-heterocyclyl, NH-aryl, NH-heteroaryl, NH-alkyl-aryl, NH-alkyl-heteroaryl, N(alkyl)2, NHC(O)-alkyl, NHC(O)-cycloalkyl, NHC(O)-heterocyclyl, NHC(O)-aryl, NHC(O)-heteroaryl, NHSO2-alkyl, NHSO2-cycloalkyl, NHSO2-aryl, NHSO2-heteroaryl, OH, O-alkyl, O-cycloalkyl, O-heterocyclyl, O-aryl, O-heteroaryl, O-alkyl-aryl, O-alkyl-heteroaryl, OC(O)-alkyl, OC(O)-cycloalkyl, OC(O)-heterocyclyl, OC(O)-aryl, OC(O)-heteroaryl, OSO2-alkyl, OSO2-cycloalkyl, OSO2-aryl, OSO2-heteroaryl, CO2H, CO2-alkyl, CO2-cycloalkyl, CO2-heterocyclyl, CO2-aryl, CO2-heteroaryl, C(O)—NH2, C(O)NH-alkyl, C(O)NH-cycloalkyl, C(O)NH-heterocyclyl, C(O)NH-aryl, C(O)NH-heteroaryl, C(O)NH-alkyl-aryl, C(O)NH-alkyl-heteroaryl, C(O)N(alkyl)2, alkyl, or aryl,
(iii) unsubstituted or substituted heterocyclyl, wherein the heterocyclyl group can be substituted with one or more, the same or different OH, O-alkyl, O-aryl, NH2, NH-alkyl, NH-aryl, alkyl or aryl,
(iv) unsubstituted or substituted aryl, wherein the aryl group can be substituted with one or more, the same or different F, Cl, Br, I, CF3, NH2, NH-alkyl, NH-cycloalkyl, NH-heterocyclyl, NH-aryl, NH-heteroaryl, NH-alkyl-cycloalkyl, NH-alkyl-heterocyclyl, NH-alkyl-aryl, NH-alkyl-heteroaryl, NH-alkyl-NH2, NH-alkyl-OH, N(alkyl)2, NHC(O)-alkyl, NHC(O)-cycloalkyl, NHC(O)-heterocyclyl, NHC(O)-aryl, NHC(O)-heteroaryl, NHSO2-alkyl, NHSO2-aryl, NHSO2-heteroaryl, NO2, SH, S-alkyl, S-cycloalkyl, S-heterocyclyl, S-aryl, S-heteroaryl, OH, OCF3, O-alkyl, O-cycloalkyl, O-heterocyclyl, O-aryl, O-heteroaryl, O-alkyl-cycloalkyl, O-alkyl-heterocyclyl, O-alkyl-aryl, O-alkyl-heteroaryl, O-alkyl-OH, O—(CH2)n—O, OC(O)-alkyl, OC(O)-cycloalkyl, OC(O)-heterocyclyl, OC(O)-aryl, OC(O)-heteroaryl, OSO2-alkyl, OSO2-cycloalkyl, OSO2-aryl, OSO2-heteroaryl, C(O)-alkyl, C(O)-aryl, C(O)-heteroaryl, CO2H, CO2-alkyl, CO2-cycloalkyl, CO2-heterocyclyl, CO2-aryl, CO2-heteroaryl, CO2-alkyl-cycloalkyl, CO2-alkyl-heterocyclyl, CO2-alkyl-aryl, CO2-alkyl-heteroaryl, C(O)—NH2, C(O)NH-alkyl, C(O)NH-cycloalkyl, C(O)NH-heterocyclyl, C(O)NH-aryl, C(O)NH-heteroaryl, C(O)NH-alkyl-cycloalkyl, C(O)NH-alkyl-heterocyclyl, C(O)NH-alkyl-aryl, C(O)NH-alkyl-heteroaryl, C(O)N(alkyl)2, C(O)N(cycloalkyl)2, C(O)N(aryl)2, C(O)N(heteroaryl)2, SO-alkyl, SO-aryl, SO2-alkyl, SO2-aryl, SO2NH2, SO2NH-alkyl, SO2NH-aryl, SO2NH-heteroaryl, SO3H, SO2O-alkyl, SO2O-aryl, SO2O-heteroaryl, alkyl, cycloalkyl, heterocyclyl, aryl or heteroaryl, and n can have the value 1, 2 or 3,
(v) unsubstituted or substituted heteroaryl, wherein the heteroaryl group can be substituted with one or more, the same or different F, Cl, Br, I, CF3, NH2, NH-alkyl, NH-cycloalkyl, NH-heterocyclyl, NH-aryl, NH-heteroaryl, NH-alkyl-aryl, NH-alkyl-heteroaryl, N(alkyl)2, NHC(O)-alkyl, NHC(O)-cycloalkyl, NHC(O)-heterocyclyl, NHC(O)-aryl, NHC(O)-heteroaryl, NHSO2-alkyl, NHSO2-aryl, NHSO2-heteroaryl, NO2, SH, S-alkyl, S-aryl, OH, OCF3, O-alkyl, O-cycloalkyl, O-heterocyclyl, O-aryl, O-heteroaryl, OC(O)-alkyl, OC(O)-cycloalkyl, OC(O)-heterocyclyl, OC(O)-aryl, OC(O)-heteroaryl, OSO2-alkyl, OSO2-cycloalkyl, OSO2-aryl, OSO2-heteroaryl, C(O)-alkyl, C(O)-aryl, C(O)-heteroaryl, CO2H, CO2-alkyl, CO2-cycloalkyl, CO2-heterocyclyl, CO2-aryl, CO2-heteroaryl, CO2-alkyl-cycloalkyl, CO2-alkyl-heterocyclyl, CO2-alkyl-aryl, CO2-alkyl-heteroaryl, C(O)—NH2, C(O)NH-alkyl, C(O)NH-cycloalkyl, C(O)NH-heterocyclyl, C(O)NH-aryl, C(O)NH-heteroaryl, C(O)NH-alkyl-cycloalkyl, C(O)NH-alkyl-heterocyclyl, C(O)NH-alkyl-aryl, C(O)NH-alkyl-heteroaryl, C(O)N(alkyl)2, C(O)N(cycloalkyl)2, C(O)N(aryl)2, C(O)N(heteroaryl)2, SO2-alkyl, SO2-aryl, SO2NH2, SO2NH-alkyl, SO2NH-aryl, SO2NH-heteroaryl, SO3H, SO2O-alkyl, SO2O-aryl, SO2O-heteroaryl, alkyl, cycloalkyl, heterocyclyl, aryl or heteroaryl;
which can be used for the production of a medicament for the treatment or prevention of physiological and/or pathophysiological states in mammals mediated by signal transduction pathways selected from the group consisting of: the “ras-Raf-Mek-Erk signal transduction pathway, the PI3K-Akt signal transduction pathway and/or the SAPK signal transduction pathway.
In a preferred embodiment, compounds according to the general formula (I) are prepared, wherein the alkyl group is selected from the group consisting of: “methyl, ethyl, n-propyl, 2-propyl, n-butyl, sec.-butyl, tert.-butyl, n-pentyl, iso-pentyl, neo-pentyl, n-hexyl, 2-hexyl, n-octyl, ethylenyl (vinyl), ethynyl, propenyl (—CH2CH═CH2; —CH═CH—CH3, —C(═CH2)-CH3), propinyl (—CH2-C≡CH, —C≡C—CH3), butenyl, butinyl, pentenyl, pentinyl, hexenyl, hexinyl, heptenyl, heptinyl, octenyl, octinyl” which can be used for the production of a medicament for the treatment or prevention of physiological and/or pathophysiological states in mammals mediated by signal transduction pathways selected from the group consisting of: the “ras-Raf-Mek-Erk signal transduction pathway, the PI3K-Akt signal transduction pathway and/or the SAPK signal transduction pathway.
In a further preferred embodiment, compounds according to the general formula (I) are prepared for the aforementioned use, wherein the heterocyclyl group is selected from the group consisting of: “tetrahydrofuryl, tetrahydropyranyl, pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl”.
In a further preferred embodiment, compounds according to the general formula (I) are prepared for the aforementioned use, wherein the heteroaryl group is selected from the group consisting of: “pyrrolyl, furyl, thienyl, thiazolyl, oxazolyl, isoxazolyl, pyrazolyl, imidazolyl, pyridinyl, pyrimidinyl, pyridazinyl, pyrazinyl, phthalazinyl, indolyl, indazolyl, indolizinyl, quinolinyl, isoquinolinyl, quinoxalinyl, quinazolinyl, carbazolyl, phenazinyl, phenothiazinyl, acridinyl”.
In a further preferred embodiment compounds according to the general formula (I) are prepared for the aforementioned use, wherein the alkyl group is selected from the group consisting of: “methyl, ethyl, n-propyl, 2-propyl, n-butyl, sec.-butyl, tert.-butyl, n-pentyl, iso-pentyl, neo-pentyl, n-hexyl, 2-hexyl, n-octyl, ethylenyl (vinyl), ethynyl, propenyl (—CH2CH═CH2; —CH═CH—CH3, —C(═CH2)-CH3), propinyl (—CH2-C≡CH, —C≡C—CH3), butenyl, butinyl, pentenyl, pentinyl, hexenyl, hexinyl, heptenyl, heptinyl, octenyl, octinyl” and/or wherein the heterocyclyl group is selected from the group consisting of: “tetrahydrofuryl, tetrahydropyranyl, pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl” and/or the heteroaryl group is selected from the group consisting of: “pyrrolyl, furyl, thienyl, thiazolyl, oxazolyl, isoxazolyl, pyrazolyl, imidazolyl, pyridinyl, pyrimidinyl, pyridazinyl, pyrazinyl, phthalazinyl, indolyl, indazolyl, indolizinyl, quinolinyl, isoquinolinyl, quinoxalinyl, quinazolinyl, carbazolyl, phenazinyl, phenothiazinyl, acridinyl”.
The inventive object was surprisingly achieved in a further aspect by preparing pyridopyrazine compounds selected from the group consisting of:
Figure US08937068-20150120-C00003
Figure US08937068-20150120-C00004
Figure US08937068-20150120-C00005
Figure US08937068-20150120-C00006
Figure US08937068-20150120-C00007
Figure US08937068-20150120-C00008
Figure US08937068-20150120-C00009
Figure US08937068-20150120-C00010
Figure US08937068-20150120-C00011
Figure US08937068-20150120-C00012
Figure US08937068-20150120-C00013
Figure US08937068-20150120-C00014
Figure US08937068-20150120-C00015
Figure US08937068-20150120-C00016
Figure US08937068-20150120-C00017
Figure US08937068-20150120-C00018
Figure US08937068-20150120-C00019
Figure US08937068-20150120-C00020
Figure US08937068-20150120-C00021
Figure US08937068-20150120-C00022
Figure US08937068-20150120-C00023
Figure US08937068-20150120-C00024
Figure US08937068-20150120-C00025
Figure US08937068-20150120-C00026

which can be used for the production of a medicament for the treatment or prevention of physiological and/or pathophysiological states in mammals mediated by signal transduction pathways selected from the group consisting of: the “ras-Raf-Mek-Erk signal transduction pathway, the PI3K-Akt signal transduction pathway and/or the SAPK signal transduction pathway.
In order to avoid ambiguities: when chemical structure and chemical name of the explicit compounds shown above erroneously do not match one another, the chemical structure shall unambiguously define the particular explicit compound.
The afore-mentioned generic compounds having the general formula (I) and preferred embodiments as well as the explicitly specified pyridopyrazine compounds 1 to 269 are hereinafter designated jointly as “compounds according to the invention”.
The expressions and terms specified to explain the compounds according to the invention having the general formula (I), the preferred embodiments and compounds 1 to 269 basically have the following meanings unless specified otherwise in the description and the claims:
In the context of this invention, the expression “alkyl” encompasses acyclic saturated or unsaturated hydrocarbon radicals which may be branched or straight-chain and have 1 to 8 carbon atoms, i.e. C1-8-alkanyls, C2-8-alkenyls and C2-8-alkynyls. Alkenyls have at least one C—C double bond and alkynyls at least one C—C triple bond. Alkynyls may additionally also have at least one C—C double bond. Preferred alkyl radicals are methyl, ethyl, n-propyl, 2-propyl, n-butyl, sec-butyl, tert-butyl, n-pentyl, iso-pentyl, neo-pentyl, n-hexyl, 2-hexyl, n-heptyl, n-octyl, n-nonyl, n-decyl, n-undecyl, n-dodecyl, ethylenyl (vinyl), ethynyl, propenyl (—CH2CH═CH2; —CH═CH—CH3, —C(═CH2)—CH3), propynyl (—CH2—C≡CH, —C≡C—CH3), butenyl, butynyl, pentenyl, pentynyl, hexenyl, hexynyl, heptenyl, heptynyl, octenyl, octadienyl and octynyl.
For the purposes of this invention, the expression “cycloalkyl” means cyclic nonaromatic hydrocarbons having 1 to 3 rings with 3 to 20, preferably 3 to 12 carbon atoms, which may be saturated or unsaturated, more preferably (C3-C8)cycloalkyl. The cycloalkyl radical may also be part of a bi- or polycyclic system, where, for example, the cycloalkyl radical is fused to an aryl, heteroaryl or heterocyclyl radical as defined herein by any possible and desired ring member(s).
The bonding to the compounds of the general formula (I) can be effected via any possible ring member of the cycloalkyl radical. Preferred cycloalkyl radicals are cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclodecyl, cyclohexenyl, cyclopentenyl and cyclooctadienyl.
The expression “heterocyclyl” represents a 3- to 14-membered, preferably 3-, 4-, 5-, 6-, 7- or 8-membered, cyclic organic radical which contains at least 1 heteroatom, optionally 2, 3, 4 or 5 heteroatoms, especially nitrogen, oxygen and/or sulphur, the heteroatoms being the same or different and the cyclic radical being saturated or unsaturated but not aromatic. The heterocyclyl radical may also be part of a bi- or polycyclic system, where, for example, the heterocyclyl radical is fused to an aryl, heteroaryl or cycloalkyl radical as defined herein by any possible and desired ring member(s). The bonding to the compounds of the general formula (I) can be effected via any possible ring member of the heterocyclyl radical. Preferred heterocyclyl radicals are tetrahydrofuryl, pyrrolidinyl, imidazolidinyl, thiazolidinyl, tetrahydropyranyl, piperidinyl, piperazinyl, morpholinyl, thiapyrrolidinyl, oxapiperazinyl, oxapiperidinyl and oxadiazolyl.
In the context of this invention, the expression “aryl” means aromatic hydrocarbons having 3 to 14 carbon atoms, preferably 5 to 14 carbon atoms, more preferably 6 to 14 carbon atoms. The aryl radical may also be part of a bi- or polycyclic system, where, for example, the aryl radical is fused to a heterocyclyl, heteroaryl or cycloalkyl radical as defined herein by any possible and desired ring member(s), for example to tetrahydrofuran, tetrahydrothiophene, pyrrolidine, imidazolidine, thiazolidine, tetrahydropyran, dihydropyran, piperidine, furan, thiophene, imidazole, thiazole, oxazole, isoxazole. The bonding to the compounds of the general formula (I) can be effected via any possible ring member of the aryl radical. Preferred aryl radicals are phenyl, biphenyl, naphthyl and anthracenyl, but likewise indanyl, indenyl or 1,2,3,4-tetrahydronaphthyl.
The expression “heteroaryl” represents a 5-, 6- or 7-membered cyclic aromatic radical which contains at least 1 heteroatom, if appropriate also 2, 3, 4 or 5 heteroatoms, especially nitrogen, oxygen and/or sulphur, the heteroatoms being the same or different. The number of nitrogen atoms is preferably 0 to 3, that of oxygen and sulphur atoms preferably 0 or 1. The heteroaryl radical may also be part of a bi- or polycyclic system, where, for example, the heteroaryl radical is fused to a heterocyclyl, aryl or cycloalkyl radical as defined herein by any possible and desired ring member(s). The bonding to the compounds of the general formula (I) can be effected via any possible ring member of the heteroaryl radical. Preferred heteroaryl radicals are pyrrolyl, furyl, thienyl, thiazolyl, isothiazolyl, oxazolyl, oxadiazolyl, isoxazolyl, pyrazolyl, imidazolyl, triazole, tetrazole, pyridinyl, pyrimidinyl, pyridazinyl, pyrazinyl, triazinyl, phthalazinyl, indolyl, indazolyl, indolizinyl, quinolinyl, isoquinolinyl, quinoxalinyl, quinazolinyl, pteridinyl, carbazolyl, phenazinyl, phenoxazinyl, phenothiazinyl, and acridinyl.
For the purposes of the present invention, the expressions “alkyl-cycloalkyl”, “cycloalkylalkyl”, “alkyl-heterocyclyl”, “heterocyclylalkyl”, “alkyl-aryl”, “arylalkyl”, “alkyl-heteroaryl” and “heteroarylalkyl” mean that alkyl, cycloalkyl, heterocycl, aryl and heteroaryl are each as defined above, and the cycloalkyl, heterocyclyl, aryl and heteroaryl radical is bonded to the compounds of the general formula (I) via an alkyl radical, preferably C1-C8-alkyl radical, more preferably C1-C4-alkyl radical.
In connection with “alkyl”, “cycloalkyl”, “heterocyclyl”, “aryl”, “heteroaryl”, alkyl-cycloalkyl”, “alkyl-heterocyclyl”, “alkyl-aryl” and “alkyl-heteroaryl” the term substituted is understood in the sense of this invention unless defined explicitly above in the description and the claims as the substitution of one or more hydrogen groups by F, Cl, Br, I, CN, CF3, NH2, NH-alkyl, NH-aryl, N(alkyl)2, NO2, SH, S-alkyl, OH, OCF3, O-alkyl, O-aryl, OSO3H, OP(O)(OH)2, CHO, CO2H, SO3H or alkyl. The substituents can be the same or different and the substitutions can take place in any arbitrary and possible position of the alkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl group.
In the context of this invention, the expression “halogen” encompasses the halogen atoms fluorine, chlorine, bromine and iodine.
Multiply substituted groups are to be understood as those which are multiply, e.g. doubly, triply, substituted either at different or at the same atoms, for example, triply substituted at the same C atoms as in the case of CF3, —CH2CF3 or at different positions as in the case of —CH(OH)—CH═CH—CHCl2. The multiple substitution can take place with the same or different substituents.
Insofar as the compounds according to the invention have at least one centre of asymmetry, they can be present in the form of their racemates, in the form of the pure enantiomers and/or diastereomers or in the form of mixtures of these enantiomers and/or diastereomers. The mixtures can be present in any arbitrary mixture ratio of the stereoisomers.
Thus, for example, the compounds according to the invention which have one or a plurality of centres of chirality and which occur as their racemates can be separated into their optical isomers, that is enantiomers or diastereomers, by methods known per se. The separation can be performed by column separation at chiral phases or by recrystallisation from an optically active solvent or by using an optically active acid or base or by derivatisation with an optically active reagent, such as for example, an optically active alcohol and subsequent separation of the residue.
The inventive compounds may be present in the form of their double bond isomers as “pure” E or Z isomers, or in the form of mixtures of these double bond isomers.
As far as possible, the compounds according to the invention can be present in the form of tautomers.
If they possess a sufficiently basic group, such as for example, a primary, secondary or tertiary amine, the compounds according to the invention can be converted into their physiologically compatible salts using inorganic and organic acids. The pharmaceutically acceptable salts of the compounds according to the invention are preferably formed with hydrochloric acid, bromic acid, sulphuric acid, phosphoric acid, methane sulfonic acid, p-toluene sulfonic acid, carbonic acid, formic acid, acetic acid, trifluoroacetic acid, sulfoacetic acid, oxalic acid, malonic acid, maleic acid, succinic acid, tartaric acid, racemic acid, malic acid, embonic acid, mandelic acid, fumaric acid, lactic acid, citric acid, glutamic acid or asparaginic acid. The salts formed include, among others, hydrochloride, hydrobromide, sulfate, hydrogen sulfate, phosphate, methane sulfonate, tosylate, carbonate, hydrogen carbonate, formiate, acetate, triflate, sulfoacetate, oxalate, malonate, maleate, succinate, tartrate, malate, embonate, mandelate, fumarate, lactate, citrate, glutaminate and aspartate. The stoichiometry of the salts of the compounds according to the invention which are formed can be integer or non-integer multiples of one.
If they contain a sufficiently acidic group, such as the carboxy group, for example, the compounds according to the invention can be converted into their physiologically compatible salts using inorganic and organic bases. Possible inorganic bases are, for example, sodium hydroxide, potassium hydroxide, calcium hydroxide, possible organic bases are ethanol amine, diethanol amine, triethanol amine, cyclohexylamine, dibenzylethylene diamine and lysine. The stoichiometry of the salts of the compounds according to the invention which are formed can be integer or non-integer multiples of one.
Likewise preferred are solvates and in particular hydrates of the compounds according to the invention, which can be obtained, for example, by crystallisation from a solvent or from aqueous solution. In this context, one, two, three or an arbitrary number of solvate or water molecules can combine with the compounds according to the invention to form solvates and hydrates.
It is known that chemical substances form solids which are present in various states of order, which are designated as polymorphous forms or modifications. The various modifications of a polymorphous substance can differ strongly in respect of their physical properties. The compounds according to the invention can be present in various polymorphous forms, in which case certain modifications can be metastable.
The compounds according to the invention can likewise be present in the form of any prodrugs such as, for example, esters, carbonates, carbamates, ureas, amides or phosphates, wherein the actually biologically active form is only released by catabolism.
It is further known that chemical substances are converted to metabolites in the body which optionally can likewise induce the desired biological effect, possibly even in a more distinct form.
Corresponding prodrugs and metabolites of the compounds according to the invention should also be considered as pertaining to the invention.
It was now surprisingly and advantageously determined that the compounds according to the invention can act simultaneously or have a modulating or inhibiting effect on two or more signal transduction pathways or enzymes. In this context, it has been found that the compounds according to the invention can act or have a modulating or inhibiting effect with high selectivity.
Such a simultaneous, for example, dual modulation or inhibition of two or more signal transduction pathways, e.g. the ras-Raf-Mek-Erk signal pathway, the PI3K-Akt signal pathway and/or the SAPK signal pathway, more especially Erk1/Erk2 and/or PI3K and/or Jnk and/or p38, is advantageous compared with merely single modulation or inhibition of a signal transduction pathway since synergistic therapeutic effects can be brought about, such as for example, intensified apoptosis and faster and more efficient tumour regression.
The surprising advantageous effects of the compounds according to the invention allow multiple therapy approaches to be pursued in physiological and/or pathophysiological states or clinical pictures which are sensitive for the treatment or modulation of, or are mediated by, two or more signal transduction pathways.
It was further surprisingly and advantageously determined that the compounds according to the invention can also act with high selectivity or have a modulating or inhibiting effect on the PI3K-Akt signal transduction pathway or enzymes thereof and that the multiple mechanisms of action and therapy approaches described above can also be used with this signal pathway or enzymes.
It was further surprisingly and advantageously determined that the compounds according to the invention can also act with high selectivity or have a modulating or inhibiting effect on the SAPK signal transduction pathway or enzymes thereof and that the multiple mechanisms of action and therapy approaches described above can also be used with this signal pathway or enzymes.
It was further surprisingly and advantageously determined that the compounds according to the invention can also act with high selectivity or have a modulating or inhibiting effect on enzymes such as ATM, ATR, mTOR, DNA-PK and/or hSMG-1 and that the multiple mechanisms of action and therapy approaches described above can also be used with this signal pathway or enzymes.
The term “modulation” is understood according to the invention as follows: “activation, partial activation, inhibition, partial inhibition”. In this case, it is within the specialist knowledge of the average person skilled in the art to measure and determine such activation, partial activation, inhibition, partial inhibition by means of the usual methods of measurement and determination. Thus, a partial activation can be measured and determined in relation to a complete activation; likewise, a partial inhibition in relation to a complete inhibition.
The terms “inhibiting, inhibition and/or retardation” are understood as follows according to the invention: “partial or complete inhibiting, inhibition and/or retardation”. In this case, it is within the specialist knowledge of the average person skilled in the art to measure and determine such inhibiting, inhibition, and/or retardation by means of the usual methods of measurement and determination. Thus, a partial inhibiting, inhibition and/or retardation, for example, can be measured and determined in relation to a complete inhibiting, inhibition and/or retardation.
The terms “modulation” and “inhibiting, inhibition and/or retardation” in connection with “enzymes” and/or “kinases” within the scope of this invention relate both to the inactive form (enzymatically inactive) and/or active form (enzymatically active) of the respective enzyme and/or kinase. This means within the scope of this invention that the compound according to the invention can have a modulating effect on the inactive form, active form or both forms of the enzyme and/or kinase.
In a further aspect, the inventive object was surprisingly achieved by preparing the compounds according to the invention which can be used to produce a medicament for the treatment or prevention of physiological and/or pathophysiological states in mammals, wherein the treatment or prevention is effected by modulation of the signal transduction pathway or pathways selected from the group consisting of: the “ras-Raf-Mek-Erk signal transduction pathway, the PI3K-Akt signal transduction pathway and/or the SAPK signal transduction pathway.
In a further aspect, the inventive object was surprisingly achieved by preparing the compounds according to the invention which can be used to produce a medicament for the treatment or prevention of physiological and/or pathophysiological states in mammals, mediated by enzymes selected from the group consisting of: “ATM, ATR, mTOR, DNA-PK, hSMG-1”.
In a further aspect, the inventive object was surprisingly achieved by preparing the compounds according to the invention which can be used to produce a medicament for the treatment or prevention of physiological and/or pathophysiological states in mammals, wherein the treatment or prevention is effected by modulation of one or more enzymes selected from the group consisting of: “ATM, ATR, mTOR, DNA-PK, hSMG-1”.
In a preferred embodiment, the compounds according to the invention are prepared for use to produce a medicament for the treatment or prevention of physiological and/or pathophysiological states in mammals mediated by the ras-Raf-Mek-Erk signal transduction pathway, the PI3K-Akt signal transduction pathway and/or use to produce a medicament for the treatment or prevention of physiological and/or pathophysiological states in mammals, wherein the treatment or prevention is effected by modulation of the ras-Raf-Mek-Erk signal transduction pathway and the PI3K-Akt signal transduction pathway.
In a further aspect, the inventive object was surprisingly achieved by preparing the compounds according to the invention which can be used to produce a medicament for the treatment or prevention of physiological and/or pathophysiological states in mammals, mediated by the PI3K-Akt signal transduction pathway.
In a further aspect, the inventive object was surprisingly achieved by preparing the compounds according to the invention which can be used to produce a medicament for the treatment or prevention of physiological and/or pathophysiological states in mammals, wherein the treatment or prevention is effected by modulation of the PI3K-Akt signal transduction pathway.
In a preferred embodiment, the compounds according to the invention are prepared for use to produce a medicament for the treatment and/or prevention of physiological and/or pathophysiological states in mammals mediated by the SAPK signal transduction pathway and the PI3K-Akt signal transduction pathway and/or to produce a medicament for the treatment or prevention of physiological and/or pathophysiological states in mammals, wherein the treatment or prevention is effected by modulation of the SAPK signal transduction pathway and the PI3K-Akt signal transduction pathway.
In a further aspect, the inventive object was surprisingly achieved by preparing the compounds according to the invention which can be used to produce a medicament for the treatment or prevention of physiological and/or pathophysiological states in mammals, mediated by the SAPK signal transduction pathway.
In a further aspect, the inventive object was surprisingly achieved by preparing the compounds according to the invention which can be used to produce a medicament for the treatment or prevention of physiological and/or pathophysiological states in mammals, wherein the treatment or prevention is effected by modulation of the SAPK signal transduction pathway.
In a preferred embodiment, the compounds according to the invention are prepared for the uses described above, wherein the modulation of the ras-Raf-Mek-Erk signal transduction pathway is effected by modulation of one or more enzymes selected from the group consisting of: “tyrosine kinase, serine/threonine kinase, receptor tyrosine kinase, cytoplasmic tyrosine kinase, cytoplasmic serine/threonine kinase” and preferably selected from the group consisting of: “Erk, Erk1, Erk2”.
In a further preferred embodiment, the compounds according to the invention are prepared for the uses described above, wherein the modulation of the P13K-Akt signal transduction pathway is effected by modulation of one or more enzymes selected from the group consisting of: “PI3K, PI3Kalpha, PI3Kbeta, PI3Kgamma, PI3Kdelta, PI3K-C2alpha, PI3K-C2beta, PI3K-Vps34p”.
In a further preferred embodiment, the compounds according to the invention are prepared for the uses described above, wherein the modulation of the SAPK signal transduction pathway is effected by modulation of one or more enzymes selected from the group consisting of: “tyrosine kinase, serine/threonine kinase, receptor tyrosine kinase, cytoplasmic tyrosine kinase, cytoplasmic serine/threonine kinase” and is preferably selected from the group consisting of: “Jnk, Jnk1, Jnk2, Jnk3, p38, p38alpha, p38beta, p38gamma, p38delta”.
In a further aspect, the inventive object was surprisingly achieved by preparing the compounds according to the invention according to the aspects, preferred embodiments and uses described above which can be used to produce a medicament for the treatment or prevention of physiological and/or pathophysiological states in mammals, wherein the treatment or prevention is effected by modulation of two or more enzymes.
In a further preferred embodiment, the compounds according to the invention are prepared for the uses described above, wherein in the treatment or prevention effected by modulation of two or more enzymes, at least one enzyme is selected from the group consisting of: “Erk, Erk1, Erk2” and at least one enzyme is selected from the group consisting of: “PI3K, PI3Kalpha, PI3Kbeta, PI3Kgamma, PI3Kdelta, PI3K-C2alpha, PI3K-C2beta, PI3K-Vps34p”.
In a further preferred embodiment, the compounds according to the invention are prepared for the uses described above, wherein in the treatment or prevention effected by modulation of two or more enzymes at least one enzyme is selected from the group consisting of: “Jnk, Jnk1, Jnk2, Jnk3, p38, p38alpha, p38beta, p38gamma, p38delta” and at least one enzyme is selected from the group consisting of: “PI3K, PI3Kalpha, PI3Kbeta, PI3Kgamma, PI3Kdelta, PI3K-C2alpha, PI3K-C2beta, PI3K-Vps34p”.
In a further preferred embodiment, the compounds according to the invention are prepared for the uses described above, wherein in the treatment or prevention effected by modulation of two or more enzymes, at least one enzyme is selected from the group consisting of: “Erk, Erk1, Erk2” and at least one enzyme is selected from the group consisting of: “ATM, ATR, mTOR, DNA-PK, hSMG-1”.
In a further preferred embodiment, the compounds according to the invention are prepared for the uses described above, wherein in the treatment or prevention effected by modulation of two or more enzymes, at least one enzyme is selected from the group consisting of: “Jnk, Jnk1, Jnk2, Jnk3, p38, p38alpha, p38beta, p38gamma, p38delta” and at least one enzyme is selected from the group consisting of: “ATM, ATR, mTOR, DNA-PK, hSMG-1”.
In a further preferred embodiment, the compounds according to the invention are prepared for the uses described above, wherein in the treatment or prevention effected by modulation of two or more enzymes, at least one enzyme is selected from the group consisting of: “PI3K, PI3Kalpha, PI3Kbeta, PI3Kgamma, PI3Kdelta, PI3K-C2alpha, PI3K-C2beta, PI3K-Vps34p” and at least one enzyme is selected from the group consisting of: “ATM, ATR, mTOR, DNA-PK, hSMG-1”.
In a further preferred embodiment, the compounds according to the invention are prepared for the uses described above, wherein the modulation is an inhibition.
The compounds according to the invention can be administered within the scope of this invention to all known mammals, in particular, humans, for the treatment and/or prevention.
In another preferred embodiment, the compounds according to the invention are prepared for the uses described above, wherein the mammal is selected from the group consisting of: “human, domesticated animal, cattle, pet, beef cattle, cow, sheep, pig, goat, horse, pony, donkey, hinny, mule, hare, rabbit, cat, dog, guinea pig, hamster, rat, mouse” and is preferably a human.
The compounds according to the invention can be used within the scope of this invention for the treatment and/or prevention of all known physiological and/or pathophysiological states.
In a preferred embodiment, the compounds according to the invention are prepared for the uses described above, wherein the physiological and/or pathophysiological states are selected from the group consisting of: “malignant tumours, benign tumours, inflammatory diseases, inflammations, pain, rheumatic diseases, arthritic diseases, HIV infections, neurological or neurodegenerative diseases, rheumatism, arthritis, AIDS, ARC (AIDS related complex), Kaposi's sarcoma, tumours originating from the brain and/or nervous system and/or meninges, dementia, Alzheimer's disease, hyperproliferative diseases, psoriasis, endometriosis, scarring, benign prostatahyperplasia (BPH), diseases of the immune system, autoimmune diseases, immunodeficiency diseases, colon tumour, gastric tumour, intestinal tumour, pulmonary tumour, pancreatic tumour, ovarian tumour, prostatic tumour, leukaemia, melanoma, hepatic tumour, renal tumour, head tumour, throat tumour, glioma, breast tumour, uterine cancer, endometrial cancer, cervico-uterine carcinoma, brain tumour, adeno-acanthoma, cancer of the bladder, gastric tumour, colorectal tumour, oesophageal cancer, gynecological tumour, ovarian tumour, cancer of the thyroid, lymphoma, chronic leukaemia, acute leukaemia, restenosis, diabetes, diabetic nephropathy, fibrotic diseases, cystic fibrosis, malignant nephrosclerosis, thrombotic microangiopathy syndrome, organ transplant rejection, glomerulopathy, metabolic diseases, solid/fixed tumours, rheumatic arthritis, diabetic retinopathy, asthma, allergies, allergic diseases, chronic obstructive pulmonary diseases, inflammatory bowel disease, fibrosis, atherosclerosis, heart diseases, cardiovascular diseases, diseases of the myocardium, vascular diseases, angiogenetic diseases, kidney diseases, rhinitis, Grave's disease, focal ischaemia, cardiac failure, ischaemia, cardiac hypertrophia, renal failure, cardiac myocytic malfunction, high blood pressure, vasoconstriction, stroke, anaphylactic shock, platelet agglutination, skeletomuscular atrophy, obesity, overweight, glucosis homeostasis, congestive cardiac insufficiency, angina, heart attack, cardiac infarction, hyperglycaemia, hypoglycaemia, hypertension”.
In a further aspect of the present invention, the inventive object was surprisingly achieved by preparing the compounds according to the aspects, preferred embodiments and uses described above, for use for the production of a medicament for the treatment or prevention of physiological and/or pathophysiological states in mammals, wherein the medicament comprises at least one further pharmacologically active substance.
In a further aspect of the present invention, the inventive object was surprisingly achieved by preparing the compounds according to the aspects, preferred embodiments and uses described above, for use for the production of a medicament for the treatment or prevention of physiological and/or pathophysiological states in mammals, wherein the medicament is administered with at least one further pharmacologically active substance before and/or during and/or after treatment.
In a further aspect of the present invention, the inventive object was surprisingly achieved by preparing the compounds according to the aspects, preferred embodiments and uses described above, for use for the production of a medicament for the treatment or prevention of physiological and/or pathophysiological states in mammals, wherein the medicament is administered before and/or during and/or after treatment with radiation therapy and/or surgery.
The compounds according to the invention can be administered within the scope of this invention with all known pharmacologically active substances in a combination therapy as described.
In a preferred embodiment, the compounds according to the invention are prepared for the uses described above, wherein the further pharmacologically active substance is selected from the group consisting of: “DNA topoisomerase I and/or II inhibitors, DNA intercalators, alkylating agents, microtubuli destabilisors, hormone and/or growth factor receptor agonists and/or antagonists, antibodies against growth factors and their receptors, kinase inhibitors, antimetabolites”.
In a preferred embodiment, the compounds according to the invention are prepared for the uses described above, wherein the further pharmacologically active substance is selected from the group consisting of: “asparaginase, bleomycin, carboplatin, carmustin, chlorambucil, cisplatin, colaspase, cyclophosphamide, cytarabine, dacarbazine, dactinomycin, daunorubicin, doxorubicin(adriamycin), epirubicin, etoposide, 5-fluorouracil, hexamethylmelamine, hydroxurea, ifosfamide, irinotecan, leucovorin, lomustine, mechlorethamine, 6-mercaptopurine, mesna, methotrexate, mitomycin C, mitoxantrone, prednisolone, prednisone, procarbazine, raloxifene, streptozocin, tamoxifen, thioguanine, topotecan, vinblastine, vincristine, vindesine, aminoglutethimide, L-asparaginase, azathioprine, 5-azacytidine cladribine, busulfan, diethylstilbestrol, 2′,2′-difluorodeoxycytidine, docetaxel, erythrohydroxynonyladenine, ethinylestradiol, 5-fluorodeoxyuridin, 5-fluorodeoxyuridin monophosphate, fludarabine phosphate, fluoxymesterone, flutamide, hydroxyprogesterone caproate, idarubicin, interferon, medroxyprogesterone acetate, megestrol acetate, melphalan, mitotane, paclitaxel, oxaliplatin, pentostatin, N-phosphonoacetyl-L-aspartate (PALA), plicamycin, semustine, teniposide, testosterone propionate, thiotepa, trimethylmelamine, uridine, vinorelbin, epothilone, gemcitabine, Taxotere, BCNU, CCNU, DTIC, 5-fluorouarcil, Herceptin, Avastin, Erbitux, Sorafenib, Gleevec, Iressa, Tarceva, rapamycin, actinomycin D”.
In a further aspect of the present invention, the inventive object was surprisingly achieved by preparing the pyridopyrazine selected from the group consisting of:
Figure US08937068-20150120-C00027
Figure US08937068-20150120-C00028
Figure US08937068-20150120-C00029
Figure US08937068-20150120-C00030
Figure US08937068-20150120-C00031
Figure US08937068-20150120-C00032
Figure US08937068-20150120-C00033
Figure US08937068-20150120-C00034
Figure US08937068-20150120-C00035
Figure US08937068-20150120-C00036
Figure US08937068-20150120-C00037
Figure US08937068-20150120-C00038
Figure US08937068-20150120-C00039
Figure US08937068-20150120-C00040
Figure US08937068-20150120-C00041
Figure US08937068-20150120-C00042
Figure US08937068-20150120-C00043
Figure US08937068-20150120-C00044
Figure US08937068-20150120-C00045
Figure US08937068-20150120-C00046
Oral administration can take place, for example, in solid form as tablet, capsule, gel capsule, dragee, granule or powder but also in the form of a potable solution. For oral administration, the new compounds according to the invention, as defined hereinbefore, can be combined with known physiologically compatible adjuvants and excipients usually used, such as gum Arabic, talc, starch, sugar such as, for example, mannite, methyl cellulose, lactose, gelatine, surfactants, magnesium stearate, cyclodextrin, aqueous or non-aqueous excipients, diluents, dispersants, emulsifiers, lubricants, preservatives and flavourings (e.g. ether oils). The compounds according to the invention can also be dispersed in a microparticle, e.g. nanoparticle composition.
Non-oral administration can be effected, for example, by intravenous, subcutaneous or intramuscular injection of sterile aqueous or oily solutions, suspensions or emulsions, by means of implants or by ointments, creams or suppositories. Optionally, administration can be effected as a retard form. Implants can contain inert materials, e.g. biologically degradable polymers or synthetic silicones such as, for example, silicone rubber. Intravaginal administration can be effected by means of vaginal rings, for example. Intrauterine administration can take place, for example, by means of diaphragms or other suitable intrauterine devices. In addition, transdermal administration can be provided, in particular by means of a formulation suitable for this purpose and/or suitable means such as plasters, for example.
As has already been explained, the new compounds according to the invention can also be combined with further pharmaceutically active substances. Within the framework of a combination therapy, the individual active constituents can be administered simultaneously or separately and either by the same pathway (e.g. oral) or by separate pathways (e.g. oral and as injection). They can be present or administered in the same or different quantities in a unit dose. A certain dosage regime can be applied insofar as this seems appropriate. In this way, a plurality of the new compounds according to the invention can be combined with one another.
The dosage can vary according to the type of indication, the severity of the disease, the type of administration, the age, sex, body weight and sensitivity of the subject to be treated over a wide range. It is within the capabilities of a person skilled in the art to determine a “pharmacologically effective quantity” of the combined pharmaceutical composition. The administration can be made in a single dose or a plurality of separate doses.
A suitable unit dose is 0.001 mg to 100 mg of the active substance, i.e. at least one compound according to the invention and optionally a further pharmaceutically active substance, per kg body weight of a patient.
In a further aspect of the present invention, accordingly pharmaceutical compositions comprising a pharmacologically active quantity of at least one compound selected from the group consisting of: “compound 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112, 113, 114, 115, 116, 117, 118, 119, 120, 121, 122, 123, 124, 125, 126, 127, 128, 129, 130, 131, 132, 133, 134, 135, 136, 138, 139, 140, 141, 142, 143, 144, 145, 146, 147, 148, 149, 150, 151, 152, 153, 154, 155, 156, 157, 158, 159, 160, 161, 162, 163, 164, 165, 166, 167, 168, 169, 170, 171, 172, 173, 174, 175, 176, 177, 178, 179, 180, 181, 182, 183, 184, 185, 186, 187, 188, 189, 190, 191, 192, 193, 194, 195, 196, 197, 198, 199, 200, 201, 202, 203, 204, 205, 206, 207, 208, 209, 210, 211, 212, 213, 214, 215, 216, 217, 218, 219, 220, 221, 222, 223, 224, 225, 226, 227, 228, 229, 230, 231, 232, 233, 234, 235, 236, 237, 238, 239, 240, 241, 242, 243, 244, 245, 246, 247, 248, 249, 250, 251, 252, 253, 254, 255, 256, 257, 258, 259, 260, 261, 262, 263, 264, 265, 266, 267, 268 and/or compound 269” and optionally pharmaceutically compatible excipients and/or adjuvants are covered by the present invention.
Preferred and particularly preferred pharmaceutical compositions are those which comprise at least one of the aforesaid preferred compounds according to the invention. Pharmaceutical compositions according to the present invention can also contain, in addition to at least one compound according to the invention, as defined previously, at least one further pharmaceutically active substance, as has been described in detail hereinbefore.
The pharmaceutical compositions according to the invention contain at least one of the new compounds according to the invention, as defined hereinbefore, in a pharmacologically active quantity, preferably in a unit dose, e.g. the aforesaid unit dose and preferably in an administration form which allows oral administration.
With regard to pharmaceutical compositions comprising compounds according to the invention and with regard to the use of the compounds according to the invention as medicaments, reference is made to the statements made in connection with the use of the new compounds according to the invention themselves with regard to the possibilities for usage and administration.
In a further aspect of the present invention, the inventive object was surprisingly solved by preparing a kit comprising a pharmacologically active quantity of at least one preferred compound according to the invention as presented above and a pharmacologically active quantity of at least one further pharmacologically active substance as defined hereinbefore.
The naming of the compounds according to the invention having the general formula (I) together with preferred exemplary embodiments and in particular compounds 1 to 269 was made using AutoNom 2000-Software (ISIS™/Draw 2.5; MDL).
General Synthetic Regulations for the Compounds According to the Invention
The procedures for manufacturing substituted pyrido[2,3-b]pyrazine according to the invention are explained below.
The compounds according to the invention can be obtained according to the diagrams below (diagrams 1-7) or the corresponding procedures known to the person skilled in the art. In addition, refer to patent specifications WO2004/104002 and WO 2004/104003 or to the corresponding methods known in the literature to manufacture the compounds in accordance with the invention.
The residues X1 to X21 listed in the diagrams below correspond in their importance to the substituents defined above in conjunction with the general formula (I), for example, R-residues etc. The particular coordination is simple for the person skilled in the art to manage based on the average knowledge of the person skilled in the art.
Figure US08937068-20150120-C00047
Initial stages for selected examples of the pyrido[2,3-b]pyrazine according to the invention where the substituents R2 and R4 are to be replaced by hydrogen, can for example, be obtained according to the procedure in diagram 2 or a corresponding procedure known to the person skilled in the art.
Figure US08937068-20150120-C00048
Initial stages for selected examples of the pyrido[2,3-b]pyrazine according to the invention, where the substituents R3 and/or R4 are to be the residues OX3, SX4, NX5X6, can for example, be obtained according to the procedure in diagram 3 or a corresponding procedure known to the person skilled in the art.
Figure US08937068-20150120-C00049
For the initial stage shown above 11 the intermediate product 9 in diagram 2 or the intermediate product 21 also in diagram 6 or an appropriate substituted intermediate product can be used.
Substituted initial stages for selected examples of the pyrido[2,3-b]pyrazine according to the invention can, for example, be obtained according to the procedure in diagram 4 or a corresponding procedure known to the person skilled in the art.
Figure US08937068-20150120-C00050
Translation of initial stage 16 in diagram 4 into the pyrido[2,3-b]pyrazines substituted according to the invention, can, for example, take place according to the procedure in diagram 5 or a corresponding procedure known to the person skilled in the art.
Figure US08937068-20150120-C00051
For the initial stage shown above 16 the intermediate product 15 in diagram 4 can be used.
Selected examples of the pyrido[2,3-b]pyrazine according to the invention where the substituents R3 and/or R4 can be selected alkyl, aryl or hetero-aryl residues, can be obtained, for example, according to the procedure in diagram 6 or corresponding procedures known to the person skilled in the art.
Figure US08937068-20150120-C00052
When manufacturing the pyridopyrazine substituted for aryl, hetero-aryl according to the invention, for the initial phase shown above 21 the intermediate product 9 in diagram 2 or the intermediate product 11 in diagram 3 can be used or an appropriate substituted intermediate product.
Selected examples of the pyrido[2,3-b]pyrazine according to the invention, where the substituents R3 and/or R4 can be selected O, S, or N substituted residues, can for instance, be obtained according to the procedure in diagram 7 or appropriate procedures known to the person skilled in the art.
Figure US08937068-20150120-C00053
The cleavage of the corresponding phosphoric ester can be carried out according to procedures known or known to the person skilled in the art using methods known in the literature.
The initial compounds and intermediate stages can either be obtained on the market or manufactured according to procedures known to the person skilled in the art. The feed materials 4, 7, 9-16, 21, 24 and 26 represent valuable intermediate compounds for manufacturing pyridopyrazine according to the invention.
In order to manufacture the initial compounds, intermediate compounds and the pyridopyrazine according to the invention, refer amongst other things, to the primary literature below, the content of which is herewith to become an integral part of the disclosure of the present filing application:
  • 1) Houben-Weyl, Methods of Organic Chemistry, Volume 4/1a, pp. 343-350
  • 2) Houben-Weyl, Methods of Organic Chemistry, 4th edition, Volume E 7b (Part 2), p. 579; Degussa GB 1184848 (1970); p. Seko, et al. EP 735025 (1996)
  • 3) D. Catarzi, et al.; J. Med. Chem. 1996, 1330-1336; J. K. Seydel, et al.; J. Med. Chem. 1994, 3016-3022
  • 4) Houben-Weyl, Methods of Organic Chemistry, Volume E 9c, pp. 231-235
  • 5) Houben-Weyl/Science of Synthesis, Volume 16, p. 1269
  • 6) C. L. Leese, H. N. Rydon J. Chem. Soc. 1955, 303-309; T. S. Osdene, G. M. Timmis J. Chem. Soc. 1955, 2033-2035
  • 7) W. He, et al. Bioorg. Med. Chem. Lett. 2003, 13, 3097-3100
  • 8) M. S. A. El-Gaby, et al. Indian J. Chem. Sect. B 2001, 40, 195-200; M. R. Myers, et al. Bioorg. Med. Chem. Lett. 2003, 13, 3091-3096; A. R. Renslo, et al. J. Amer. Chem. Soc. 1999, 121, 7459-7460; C. O. Okafor, et al. J. Heterocyclic Chem. 1983, 20, 199-203; C. R. Hopkins, et al. Tet. Lett. 2004, 45, 8631-8633
  • 9) J. Yin, et al. Org. Lett. 2002, 4, 3481-3484; O. A. El-Sayed, et al. Arch. Pharm. 2002, 335, 403-410; C. Temple, et al. J. Med. Chem. 1992, 35, 988-993
  • 10) A. M. Thompson, et al. J. Med. Chem. 2000, 43, 4200-4211; N. A. Dales, et al. Org. Lett. 2001, 2313-2316; G. Dannhardt, et al. Arch. Pharm. 2000, 267-274; G. S. Poindexter, et al. Bioorg. Med. Chem. 2004, 12, 507-521; J.-M. Receveur, et al. Bioorg. Med. Chem. Lett. 2004, 14, 5075-5080
  • 11) G. Heinisch, et al. Arch. Pharm. 1997, 207-210; K. Matsuno, et al. J. Med. Chem. 2002, 45, 4513-4523; A. M. Papini, et al. J. Med. Chem. 2004, 47, 5224-5229
  • 12) L. Mao, et al. Synthesis 2004, 15, 2535-2539; M. Darabantu, et al. Tetrahedron 2005, 61, 2897-2905; E. Ford, et al. Tet. Lett. 2000, 41, 3197-3198; T. Shiota, et al. J. Org. Chem. 1999, 64, 453-457
  • 13) J. F. Miravet, et al. Org. Lett. 2005, 7, 4791-4794; A. L. Castelhano, et al. Bioorg. Med. Chem. Lett. 2005, 15, 1501-1504.
  • 14) J. W. Huffmann, et al. Bioorg. Med. Chem. 2006, 14, 247-262; T. Liu, et al. Org. & Biomolecular Chem. 2005, 3, 1525-1533
    General Regulations Setting Out the Compounds According to the Invention:
    Diagram 1: Stage 1
2,6-diamino-3-nitropyridine or 2-amino-3,5-dinitro-pyridine is dissolved in a suitable inert solvent, such as methanol, ethanol, dimethylformamide or dioxan, for instance. After adding a catalyst, Raney nickel. palladium on carbon or platinum(IV) oxide, the reaction mass is placed in a hydrogen atmosphere with pressure of between 1 and 5 bar being set. The reaction mass is allowed to react for several hours, 1-16 hours, for example, in a temperature range between 20° C. and 60° C. When conversion is finished, the insoluble residues are filtered out, the filter medium can, for example, consist of silica gel, celite or normal glass fibre filters and rewashed using the corresponding solvent. The raw product present in solution is used for the next conversion without additional purification.
Stage 2
The 1,2-dione derivative is placed in a suitable, inert solvent, methanol, ethanol, dioxan, toluene or dimethylformamide. Immediately after reduction 2,3,6-triaminopyridine or 2,3,5-triaminopyridine is added as solution of the raw products in one of the solvents referred to above to the presented 1,2-dione, if necessary adding an acid, such as for example, acetic acid or a base, potassium hydroxide. The reaction mass is allowed to react for a period, between 20 minutes and 40 hours in a temperature range of between 20° C. and 80° C. Once conversion is finished, a possible precipitate which has separated is filtered out, the filter medium can, for instance, consist of filter paper on the market, rewashed with the corresponding solvent and the remaining solid is dried in vacuum or rather the reaction mass is released from the solvent in vacuum. Where dimethylformamide is used, the reaction mass is stirred into a large quantity of water and the precipitate separated is filtered out or rather the aqueous phase is extracted using a suitable organic solvent, such as, for example, dichloromethane or ethyl acetate and the organic phases concentrated in vacuum. Cleaning of the remaining raw product takes place by recrystallisation from a suitable solvent, dioxan, for example, or using column or rather flash chromatography on silica gel or aluminium oxide. A mixture of methanol and dichloromethane, for example, can serve as solvent.
Diagram 2: Stage 1
The pyrido-pyrazine on derivate 8 is presented in a suitable, inert solvent, dimethylformamide, dioxan or toluene or without a solvent. A chlorinating agent, phosphoryl chloride or thionyl chloride, for instance, is added at room temperature and the reaction amount is allowed to react in a temperature range of between 20° C. and 100° C. for a period, between 1 hour and 24 hours for example. When conversion is finished, the reaction mass is poured on to water and neutralised using a suitable aqueous base, caustic soda solution, for instance. A possible precipitate separated is filtered out, the filter medium can consist, for instance of filter paper on the market, is rewashed with the corresponding solvent and the remaining solid dried in vacuum or the aqueous phase is extracted in a suitable organic solvent, such as dichloromethane or ethyl acetate, for example, and the organic phases are concentrated. Cleaning of the remaining raw product takes place by recrystallisation from a suitable solvent, dioxan or toluene, for example, or using column or rather flash chromatography on silica gel or aluminium oxide. A mixture of methanol and dichloromethane, for example, can serve as solvent.
Diagram 3: Stage 1
2,3,6-triaminopyridine or 2,3,5-triaminopyridine are presented directly after reduction as a solution of the raw products in one of the solvents referred to above. Having added an oxalic acid derivative, such as oxalic acid diethylester or oxalyl chloride, for example, the reaction mass is allowed to react, if necessary by adding an acid, such as hydrochloric acid, sulphuric acid or crystalline acid for a period of time, between 10 minutes and 24 hours, for instance, in a temperature range of between 20° C. and 150° C. Once conversion is finished, a possible precipitate which has separated is filtered out, the filter medium can, for instance, consist of filter paper on the market, rewashed with the corresponding solvent and the remaining solid is dried in vacuum or rather the reaction mass is released from the solvent in vacuum. Alternatively, the reaction mass can be stirred into a large quantity of water and the precipitate separated filtered out or rather the aqueous phase, following neutralisation using an appropriate aqueous base, such as for instance, caustic soda solution, extracted using a suitable organic solvent, such as for instance, dichloromethane or ethyl acetate, and the organic phase concentrated in vacuum. Cleaning of the remaining raw product takes place by recrystallisation from a suitable solvent, dioxan or toluene, for example, or using column or rather flash chromatography on silica gel or aluminium oxide. A mixture of methanol and dichloromethane, for example, can serve as solvent.
Stage 2
The diketone derivative 10 is presented in a suitable, inert solvent, dimethylformamide, dioxan or toluene, for instance, or without a solvent. A chlorinating agent, phosphoryl chloride or thionyl chloride, for instance, is added at room temperature and the reaction amount is allowed to react in a temperature range of between 20° C. and 100° C. for a period, between 1 hour and 24 hours, for example. When conversion is finished, the reaction mass is poured on to water and neutralised using a suitable aqueous base, caustic soda solution, for instance. A possible precipitate separated is filtered out, the filter medium can consist, for instance of filter paper on the market, is rewashed with the corresponding solvent and the remaining solid dried in vacuum or the aqueous phase is extracted in a suitable organic solvent, such as dichloromethane or ethyl acetate, for example, and the organic phases are concentrated in vacuum. Cleaning of the remaining raw product takes place by recrystallisation from a suitable solvent, dioxan or toluene, for example, or using column or flash chromatography on silica gel or aluminium oxide. A mixture of methanol and dichloromethane, for example, can serve as a solvent.
Stage 3
The intermediate stage 11 can be converted using a corresponding alcohol, thiol or amine and if necessary using a suitable base, preferably sodium hydride, pyridine, triethylamine, potassium carbonate or sodium methanolate in methanol, in a suitable, inert solvent, such as dimethylformamide, dimethyl sulfoxide, methanol, toluene or in a base as solvent, such as for example, pyridine or triethylamine without solvent. The reaction mass is allowed to react for a period, between 30 minutes and 2 days, for example, in a temperature range between 20° C. and 140° C. Alternatively, the intermediate stage 11 can be converted using a corresponding amine and a suitable catalyst, such as for instance, tris(dibenzalacetone) palladium(0) or tetrakis(triphenylphosphine) palladium(0) and a suitable ligand, such as for instance, 2-(dicyclohexylphosphanyl) biphenyl or BINAP, and a suitable base, sodium tert butanolate or potassium carbonate, in a suitable solvent, such as toluene, dioxan or dimethylformamide, for instance. The reaction mass is allowed to react for a period, between 2 hours and 30 hours, for example, in a temperature range of between 60° C. and 120° C. Once conversion is finished, a possible precipitate which has separated is filtered out, the filter medium can, for instance, consist of filter paper on the market, rewashed with the corresponding solvent and the remaining solid is dried in vacuum or the reaction mass is released from the solvent in vacuum. Alternatively, the reaction mass can be stirred into a large quantity of water and the separated precipitate filtered out or the aqueous phase, following neutralisation using a suitable aqueous acid, such as for instance, caustic soda solution, extracted using a suitable organic solvent, such as for example, dichloromethane or ethyl acetate, and the organic phase concentrated in vacuum. Cleaning of the remaining raw product takes place by recrystallisation from a suitable solvent, dioxan or toluene, for example, or using column or flash chromatography on silica gel or aluminium oxide. A mixture of methanol and dichloromethane, for example, can serve as solvent.
Diagram 4: Stage 1
The intermediate stage 15 can be converted in the corresponding suitable chloride, bromide, Iodide or tosylate and if necessary using a suitable base, preferably sodium hydride, pyridine, triethylamine, sodium carbonate or sodium methanolate in methanol in a suitable, inert solvent, such as, dimethylformamide, dimethyl sulfoxide, methanol or in a base as solvent, such as, for example, pyridine or triethylamine without a solvent. The reaction mass is allowed to react for a period, between 1 hour and 24 hours for example, in a temperature range between 20° C. and 150° C. Alternatively, the intermediate stage 15 can be converted using a corresponding aryl bromide or aryl Iodide and a suitable catalyst, such as, for example, tris(dibenzalacetone)palladium(0) or tetrakis(triphenylphosphine) palladium(0) and a suitable ligand, such as for example, 2-(dicyclohexylphosphanyl) biphenyl or BINAP, and a suitable base, for example, potassium carbonate or sodium tert butanolate or in a suitable solvent, such as toluene, dioxan or dimethylformamide, for instance. The reaction mass is allowed to react for a period, between 10 hours and 30 hours, for example, in a temperature range of between 60° C. and 120° C. Once conversion is finished, a possible precipitate separated is filtered out, the filter medium can, for instance, consist of filter paper on the market, rewashed with the corresponding solvent and the remaining solid is dried in vacuum or the reaction mass is released from the solvent in vacuum. Alternatively, the reaction mass can be stirred into a large quantity of water and the precipitate separated filtered out or the aqueous phase, following neutralisation using an suitable aqueous acid, such as for example, caustic soda solution, extracted using a suitable organic solvent, such as, for example, dichloromethane or ethyl acetate, and the organic phase concentrated in vacuum. Cleaning of the remaining raw product takes place by recrystallisation from a suitable solvent, dioxan or toluene, for example, or using column or flash chromatography on silica gel or aluminium oxide. A mixture of methanol and dichloromethane, for example, can serve as solvent.
Diagram 5: Stage 1
After the basic procedure, the products which have come about according to the basic procedure can be translated in consecutive reactions into resultant products according to the invention in a procedure known to the person skilled in the art.
Thus, if the product is to be a derivative of compound 17 according to diagram 5, then the reaction product 16 can be converted using a corresponding isocyanate and if necessary a suitable base, preferably sodium hydride, potassium hexamethyldisalzide, pyridine, triethylamine or potassium carbonate, in a suitable, inert solvent, such as dimethylformamide, dimethyl sulfoxide, acetonitrile, dichloromethane, 1,2-dichloroethane or dioxan or in a base as solvent, such as for example, pyridine or triethylamine without solvent. The reaction mass is allowed to react for several hours, between 1 and 24 hours, for instance, in a temperature range between 0 and 80° C. Once conversion is finished, a possible precipitate which has separated is filtered out, the filter medium can, for instance, consist of filter paper on the market, rewashed with the appropriate solvent and the remaining solid is dried in vacuum or the reaction mass is released from the solvent in vacuum. Alternatively, the reaction mass can be stirred into a large quantity of water and the precipitate separated filtered out or the aqueous phase, following neutralisation using an appropriate aqueous acid, such as for example, caustic soda solution, extracted using a suitable organic solvent, such as, for instance, dichloromethane or ethyl acetate, and the organic phase concentrated in vacuum. Cleaning of the remaining raw product takes place by recrystallisation from a suitable solvent, dioxan or toluene, for example, or using column or flash chromatography on silica gel or aluminium oxide. A mixture of methanol and dichloromethane, for example, can serve as a solvent.
Or alternatively, if the product is to be a derivative of the compound 18 according to diagram 5, the reaction product 16 can be converted using phosgene or carbonyldiimidazole and a corresponding amine in a suitable, inert solvent, such as, for example, dimethylformamide, tetrahydrofuran, toluene, dichloromethane or acetonitrile. If necessary, a suitable base is used, preferably pyridine, sodium hyrdogencarbonate, triethylamine, N-methylmorpholine or sodium acetate. The reaction mass is allowed to react for a period, for example between 15 minutes and 24 hours, in a temperature range between 0 and 60° C. Alternatively, the reaction product 16 can be converted using a corresponding amine phenyl carbamate reagent and if necessary using a suitable base, preferably pyridine, sodium carbonate, triethylamine or sodium hydride, in a suitable, inert solvent, such as, for example, pyridine or triethylamine or without solvent. The reaction mass is allowed to react for a period, between 1 hour and 18 hours for example, in a temperature range between 0° C. and 120° C. Once conversion is finished, a possible precipitate which has separated is filtered out, the filter medium can, for instance, consist of filter paper on the market, rewashed with the corresponding solvent and the remaining solid is dried in vacuum or the reaction mass is released from the solvent in vacuum. Alternatively, the reaction mass can be stirred into a large quantity of water and the precipitate separated is filtered out or the aqueous phase, following neutralisation using a suitable aqueous acid, such as for example, caustic soda solution, extracted using a suitable organic solvent, such as, for instance, dichloromethane or ethyl acetate, and the organic phase concentrated in vacuum. Cleaning of the remaining raw product takes place by recrystallisation from a suitable solvent, dioxan or toluene, for example, or using column or flash chromatography on silica gel or aluminium oxide. A mixture of methanol and dichloromethane, for example, can serve as solvent.
Thus if the product is to be a derivative of the compound 19 according to diagram 5, the reaction product 16 can be translated using a corresponding isothiocyanate, and if necessary, a suitable base, preferably sodium hydride, triethylamine, potassium carbonate or pyridine, in a suitable, inert solvent, such as for instance dimethylformamide, tetrahydrofuran, acetone or toluene, or in a base as solvent, such as, for instance, pyridine or triethylamine or without solvent. The reaction mass is allowed to react for a period, for example between 30 minutes and 90 hours, in a temperature range between 0 and 115° C., Once conversion is finished, a possible precipitate which has separated is filtered out, the filter medium can, for instance, consist of filter paper on the market, rewashed with the corresponding solvent and the remaining solid is dried in vacuum or the reaction mass is released from the solvent in vacuum. Alternatively, the reaction mass can be stirred into a large quantity of water and the precipitate separated filtered out or the aqueous phase, following neutralisation using a suitable aqueous acid, such as for example, caustic soda solution, extracted using a suitable organic solvent, such as, for instance, dichloromethane or ethyl acetate, and the organic phase concentrated in vacuum. Cleaning of the remaining raw product takes place by recrystallisation from a suitable solvent, dioxan or toluene, for example, or using column or flash chromatography on silica gel or aluminium oxide. A mixture of methanol and dichloromethane, for example, can serve as a solvent.
Or alternatively, if the product is to be a derivative of the compound 20 according to diagram 5, the reaction product 16 can be translated using thiophosgene or thiocarbonyl diimidazole and a corresponding amine in a suitable, inert solvent, such as dimethylformamide, tetrahydrofuran, toluene, dichloromethane, ethanol or acetonitrile. If necessary, a suitable base, preferably pyridine, sodium hydrogencarbonate, potassium carbonate, triethylamine or imidazole is used. The reaction mixture is allowed to react for several hours, between 1 and 24 hours for example, in a temperature range between −10 and 80° C. Once conversion is finished, a possible precipitate which has separated is filtered out, the filter medium can, for instance, consist of filter paper on the market, rewashed with the appropriate solvent and the remaining solid is dried in vacuum or the reaction mass is released from the solvent in vacuum. Alternatively, the reaction mass can be stirred into a large quantity of water and the precipitate separated filtered out or the aqueous phase, following neutralisation using an appropriate aqueous acid, such as for example, caustic soda solution, extracted using a suitable organic solvent, such as, for instance, dichloromethane or ethyl acetate, and the organic phase concentrated in vacuum. Cleaning of the remaining raw product takes place by recrystallisation from a suitable solvent, dioxan or toluene, for example, or using column or flash chromatography on silica gel or aluminium oxide. A mixture of methanol and dichloromethane, for example, can serve as solvent.
Diagram 6: Stage 1
After the basic procedure, the products which have come about according to the basic procedure can be translated in consecutive reactions into resultant products according to the invention in a procedure known to the person skilled in the art.
Thus, if the product is to be a derivative of the compound 22 according to diagram 6, the reaction product 21 can be translated using the corresponding aryl/heteroaryl boronic acid derivatives or aryl/heteroaryl organotin compounds and a suitable catalyst, such as, for example, Pd(PPh3)4, [1,1′-bis(diphenylphosphino)ferrocene]dichloro palladium(II) or Pd2(dba)3 and a suitable base, for example, sodium carbonate, cesium carbonate or triethylamine, in a suitable solvent, such as for instance, dimethylformamide, dimethylformamide/water, toluene, acetonitrile, dimethoxyethane or dioxan. The reaction mixture is allowed to react for a period, for instance between 6 hours and several days, in a temperature range between 60° C. and 120° C. Once conversion is finished, a possible precipitate which has separated is filtered out, the filter medium can, for instance, consist of filter paper on the market, rewashed with the corresponding solvent and the remaining solid is dried in vacuum or the reaction mass is released from the solvent in vacuum. Alternatively, the reaction mass can be stirred into a large quantity of water and the precipitate separated filtered out or the aqueous phase, following neutralisation using a suitable aqueous acid, such as for example, caustic soda solution, extracted using a suitable organic solvent, such as, for instance, dichloromethane or ethyl acetate, and the organic phase concentrated in vacuum. Cleaning of the remaining raw product takes place by recrystallisation from a suitable solvent, dioxan or toluene, for example, or using column or flash chromatography on silica gel or aluminium oxide. A mixture of methanol and dichloromethane, for example, can serve as a solvent.
Thus if the product is to be a derivative of the compound 23 according to diagram 6, the reaction product 21 can be converted using corresponding alkyl zinc halogenides and a suitable catalyst, such as, for example, Pd(PPh3)4, [1,1′-bis(diphenylphosphino)ferrocene]dichloro palladium(II) or PdCl2(PPh3)2 in a suitable solvent, such as for instance, dimethylformamide, tetrahydrofuran, toluene, dimethoxyethane or dioxan. The reaction mixture is allowed to react for a period, for example between 30 minutes and 48 hours, in a temperature range between room temperature and 120° C. Once conversion is finished, a possible precipitate which has separated is filtered out, the filter medium can, for instance, consist of filter paper on the market, rewashed with the appropriate solvent and the remaining solid is dried in vacuum or the reaction mass is released from the solvent in vacuum. Alternatively, the reaction mass can be stirred into a large quantity of water and the perpetrate separated is filtered out or the aqueous phase, following neutralisation using an appropriate aqueous acid, such as for example, dichloromethane or ethyl acetate, and the organic phase concentrated in vacuum. Cleaning of the remaining raw product takes place by recrystallisation from a suitable solvent, dioxan or toluene, for example, or using column or flash chromatography on silica gel or aluminium oxide. A mixture of methanol and dichloromethane, for example, can serve as solvent.
Diagram 7: Stage 1
After the basic procedure, the products which have come about according to the basic procedure can be translated in consecutive reactions into resultant products according to the invention in a procedure known to the person skilled in the art.
Thus, if the product is to be a derivative of the compound 25 according to diagram 7, the reaction product 24 can be converted, for example, using a corresponding chloride, bromide, Iodide and if necessary using a suitable base, preferably sodium hydride, pyridine, triethylamine, sodium carbonate or sodium methanolate in methanol in a suitable, inert solvent, such as, dimethylformamide, dimethyl sulfoxide, methanol, dioxan, tetrahydrofuran, toluene or in a base as solvent, such as, for instance, pyridine or triethylamine or without solvent. The reaction mass is allowed to react for a period, between 30 minutes and 2 days, for example, in a temperature range between 0° C. and 140° C. Alternatively, an amino substituted intermediate stage 24, for example, can be used, using a corresponding chloride, bromide or Iodide and a suitable catalyst, such as, for instance, tris(dibenzalacetone)palladium(0) or tetrakis(triphenylphosphine) palladium(0) and a suitable ligand, such as, for instance, 2-(dicyclohexylphosphanyl) biphenyl or BINAP, and a suitable base, sodium tert butanolate or potassium carbonate, in a suitable solvent, such as toluene, dioxan or dimethylformamide, for instance. The reaction mass is allowed to react for a period, between 2 hours and 30 hours, for example, in a temperature range of between 60° C. and 120° C. Once conversion is finished, a possible precipitate separated is filtered out, the filter medium can, for instance, consist of filter paper on the market, rewashed with the suitable solvent and the remaining solid is dried in vacuum or the reaction mass is released from the solvent in vacuum. Alternatively, the reaction mass can be stirred into a large quantity of water and the perpetrate separated filtered out or the aqueous phase, following neutralisation using a suitable aqueous acid, such as for instance, caustic soda solution, extracted using a suitable organic solvent, such as, for example, dichloromethane or ethyl acetate, and the organic phase concentrated in vacuum. Cleaning of the remaining raw product takes place by recrystallisation from a suitable solvent, dioxan or toluene, for example, or using column or flash chromatography on silica gel or aluminium oxide. A mixture of methanol and dichloromethane, for example, can serve as solvent.
Thus, if the product is to be a derivative of the compound 27 according to diagram 7, the reaction product 26 can be converted, for example, using a corresponding chloro-phosphoric ester and if necessary using a suitable base, preferably sodium hydride, pyridine, triethylamine, potassium carbonate or lithium diisopropylamide in a suitable, inert solvent, such as for example, dimethylformamide, dimethyl sulfoxide, methanol, dioxan, tetrahydrofuran, toluene or in a base as solvent, such as for example, pyridine or triethylamine or without solvent. The reaction mass is allowed to react for a period, between 1 hour and 24 hours for example, in a temperature range between 0° C. and 100° C. Once conversion is finished, a possible precipitate separated is filtered out, the filter medium can, for instance, consist of filter paper on the market, rewashed with the suitable solvent and the remaining solid is dried in vacuum or the reaction mass is released from the solvent in vacuum. Alternatively, the reaction mass can be stirred into a large quantity of water and the precipitate separated filtered out or the aqueous phase, following neutralisation using a suitable aqueous acid, such as for example, caustic soda solution, extracted using a suitable organic solvent, such as, for example, dichloromethane or ethyl acetate, and the organic phase concentrated in vacuum. Cleaning of the remaining raw product takes place by recrystallisation from a suitable solvent, dioxan or toluene, for example, or using column or rather flash chromatography on silica gel or aluminium oxide. A mixture of methanol and dichloromethane, for example, can serve as solvent.
Under the reaction conditions mentioned, OH, SH and NH2 groups can possibly undergo unwanted secondary reactions. It is therefore preferable to provide them with protective groups or in the case of NH2 to replace with NO2 and subsequently to split the protective group off or to reduce the NO2 group. Thus varying the procedures described above, in the initial compounds at least one OH group, for example, can be replaced by a benzyl oxy group and/or at least one SH group, for example, by an S-benzyl group and/or at least one NH2 group by a NH-benzyl group or by an NO2group. Subsequently at least one—preferably all—benzyl oxy group(s) or NH benzyl group(s) can be split off, for instance, using hydrogen and palladium on carbon and/or at least one—preferably all—S-benzyl group(s), for example, using sodium in ammonia and/or at least one—preferably all NO2 group(s) are reduced, for instance using hydrogen and Raney nickel to NH2
Under some of the reaction conditions mentioned OH, NH2 and COOH groups can possibly undergo unwanted secondary reactions. It is therefore preferable to convert initial compounds and intermediate stages containing at least one OH and/or at least one NH2 and/or at least one COOH group into corresponding carbonic ester and carboxamide derivatives. Thus varying the procedures described above, in the initial compounds and intermediate stages with at least one OH group and/or at least one NH2group can be converted into carboxamide derivatives by transformation using an active carboxamide group. Thus varying the procedures described above the initial compounds and intermediate stages with at least one COOH group can be converted into carbonic ester or carboxamide derivatives by transformation using an activator, such as thionyl chloride or carbonyldiimidazole and a subsequent transformation using a suitable alcohol or amine. Subsequently at least one—preferably all—carbonic ester or carboxamide group(s) in the initial compounds and intermediate stages can be split, for instance using diluted aqueous acids or bases to free at least one—preferably all —OH group(s) and/or NH2 group(s) and/or COOH groups.
The invention will be explained in detail with reference to the following examples without being restricted to these examples.
EXAMPLES I. Manufacture of Compounds According to the Invention
According to the general synthesis regulations on which the synthesis diagrams 1-7 are based, the following compounds according to the invention were synthesised.
The initial stages for the manufacture of the compounds according to the invention can be synthesised according to procedures known to the person skilled in the art—unless otherwise described.
The chemicals and solvents used were purchased commercially from the usual suppliers (Acros, Aldrich, Fluka, Lancaster, Maybridge, Merck, Sigma, TCI, etc.) or synthesised.
Example 1 1-ethyl-3-[3-(3-hydroxy-phenyl)-pyrido[2,3-b]pyrazine-6-yl]-urea (Compound 151) Manufacture of 1-(3-chloropyrido[2,3-b]pyrazine-6-yl)-3-ethyl-urea (Conversion According to Diagram 5)
100 mg 3-chloropyrido[2,3-b]pyrazine-6-yl-amine (0.55 mmol) was presented in 5 ml predried pyridine and add 44 μL ethyl isocyanate (0.55 mmol) at room temperature. The mixture was stirred at 75° C. and then a total of 132 μL ethyl isocyanate (1.65 mmol) was added again over 18 hours to the reaction mixture in small amounts. Then the volatile components were removed in vacuum. The resulting solid was cleaned through column chromatography on silica gel (solvent dichloromethane/methanol). A light yellow solid was obtained.
Refer to the following literature and the procedures known to the person skilled in the art to manufacture 3-chloropyrido[2,3-b]pyrazine-6-yl-amine:
  • 1) T. S. Osdene, et al. J. Chem. Soc. 1955, 2033-2035; C. L. Leese, et al. J. Chem. Soc. 1955, 303-309
  • 2) C. M. Atkinson, et al. J. Chem. Soc. 1956, 26-30; S. Goswami, et al. Molecules 2005, 10, 929-936
Manufacture of 1-ethyl-3-[3-(3-hydroxy-phenyl)-pyrido[2,3-b]pyrazine-6-yl]-urea (Conversion According to Diagram 6)
100 mg 1-(3-chloropyrido[2,3-b]pyrazine-6-yl)-3-ethyl-urea (0.40 mmol) was presented in a dimethylformamide/water mixture under nitrogen as protective gas. Then 60.3 mg 3-hydroxyphenyl boronic acid (0.44 mmol), 126 mg sodium carbonate (1.19 mmol) and 23 mg tetrakis(triphenylphosphine) palladium(0) (0.02 mmol) were added. The reaction mixture was stirred at 90° C. for 4 hours. The mixture was filled with water for reprocessing and the deposited solid rewashed with dichloromethane. The resulting raw product was stirred again in warm DCM, filtered off and dried. A yellow solid was obtained.
Additional compounds according to the invention were produced according to this regulation: For example, the compounds 152, 153, 154, 208, 209 and 210.
Example 2 1-ethyl-3-(3-o-tolylamino-pyrido[2,3-b]-pyrazine-6-yl)-urea (Compound 160) Manufacture of 1-ethyl-3-(3-o-tolylamino-pyrido[2,3-b]-pyrazine-6-yl)-urea (Conversion According to Diagram 3)
55 mg 1-(3-chlor-pyrido[2,3-b]pyrazine-6-yl)-3-ethyl-urea (0.22 mmol), 60 mg o-toluidine (0.56 mmol), 33 mg sodium-tert.-butylate(0.33 mmol), 29 mg tris(dibenzalacetone)palladium(0) (0.03 mmol) and 78 mg 2-(dicyclohexylphosphanyl)biphenyl (0.22 mmol) were presented in 1.5 ml predried toluene in a microwave reaction vessel under nitrogen as protective gas. The reaction mass was heated in the microwave at 100° C. (100 watt) for 30 minutes. Then the volatile components were removed in vacuum and the raw product was cleaned using column chromatography on silica gel (solvent dichloromethane/methanol). A yellow solid was obtained.
Additional compounds according to the invention were produced according to this regulation: For example, the compounds 161, 180, 211, 212 and 213.
Example 3 1-ethyl-3-(3-phenoxy-pyrido[2,3-b]pyrazine-6-yl)-urea (Compound 64) Manufacture of 1-ethyl-3-(3-phenoxy-pyrido[2,3-b]pyrazine-6-yl)-urea (Conversion According to Diagram 3)
19 mg sodium hydride (0.48 mmol) (60% suspension in mineral oil) was presented in 10 ml predried dimethylformamide. 37 mg phenol (0.40 mmol) was added dissolved in a little predried dimethylformamide at 0° C. The mixture was stirred for 1 hour at 0° C. Then 100 mg 1-(3-chloro-pyrido[2,3-b]pyrazine-6-yl)-3-ethyl-urea (0.40 mmol) was dissolved in a little predried dimethylformamide and dropped in at 0° C. The reaction mixture was stirred overnight at room temperature. When the reaction was finished the reaction solution was poured on iced water and neutralised using 1N HCl. The raw product deposited was cleaned using column chromatography on silica gel (dichloromethane/methanol). A white solid was obtained.
Additional compounds according to the invention were produced according to this regulation: For example, the compounds 67, 69, 73, 80 and 87.
Conversion using amines can be undertaken using triethylamine as a base in dioxan or without a base in dioxan. Additional compounds according to the invention were produced according to this method: For example, the compounds 59, 62, 65, 81 and 88.
II. Physico-Chemical Characterisation
II.1 ESI MS Data for Selected Compounds
TABLE 1
New pyrido[2,3-b]pyrazine derivatives with relevant MS data according to the
general formula (I)
MS m/z
Comp. Pyridopyrazine derivative (M + H+)
1 1-Allyl-3-(3-phenyl-pyrido[2,3-b]pyrazin-6-yl)-thiourea 322.0
17 1-(2-Morpholin-4-yl-ethyl)-3-(3-phenyl-pyrido[2,3-b]pyrazin-6-yl)-urea 379.1
29 1-[3-(4-Hydroxy-phenyl)-pyrido[2,3-b]pyrazin-6-yl]-3-prop-2-inyl- 336.2
thiourea
31 1-[3-(4-Hydroxy-phenyl)-pyrido[2,3-b]pyrazin-6-yl]-3-((E)-propenyl)- 338.3
thiourea
35 1-Phenethyl-3-(3-phenyl-pyrido[2,3-b]pyrazin-6-yl)-urea 370.2
36 1-(2,3-Di-pyridin-2-yl-pyrido[2,3-b]pyrazin-6-yl)-3-ethyl-urea 372.1
37 1-(2,3-Dimethyl-pyrido[2,3-b]pyrazin-6-yl)-3-ethyl-urea 246.2
38 1-[3-(4-Hydroxy-phenyl)-pyrido[2,3-b]pyrazin-6-yl]-3-methyl-urea 296.1
39 1-Allyl-3-[3-(4-phenoxy-phenyl)-pyrido[2,3-b]pyrazin-6-yl]-urea 398.1
40 Methansulfonic acid-4-[6-(3-allyl-ureido)-pyrido[2,3-b]pyrazin-3-yl]- 400.0
phenyl-ester
41 4-Dimethylamino-benzoic acid-4-[6-(3-allyl-ureido)-pyrido[2,3- 469.3
b]pyrazin-3-yl]-phenyl-ester
42 Acetic acid-4-[6-(3-allyl-ureido)-pyrido[2,3-b]pyrazin-3-yl]-phenyl- 364.1
ester
43 1-Ethyl-3-[3-(4-hydroxy-phenyl)-pyrido[2,3-b]pyrazin-6-yl]-urea 310.2
44 1-[3-(4-Amino-phenyl)-pyrido[2,3-b]pyrazin-6-yl]-3-methyl-thiourea 311.1
45 1-Ethyl-3-(3-phenyl-pyrido[2,3-b]pyrazin-6-yl)-urea 294.2
46 1-Acetyl-1-ethyl-3-(3-phenyl-pyrido[2,3-b]pyrazin-6-yl)-urea 336.3
47 1-Allyl-3-[3-(4-fluoroo-phenyl)-pyrido[2,3-b]pyrazin-6-yl]-urea 324.0
48 1,1-Diethyl-3-(3-phenyl-pyrido[2,3-b]pyrazin-6-yl)-urea 322.0
49 1-(2-Chloro-ethyl)-3-[3-(4-fluoro-phenyl)-pyrido[2,3-b]pyrazin-6-yl]- 346.3
urea
50 1-Ethyl-3-[3-(4-methoxy-phenyl)-pyrido[2,3-b]pyrazin-6-yl]-urea 324.2
51 1-(3-Phenyl-pyrido[2,3-b]pyrazin-6-yl)-3-propyl-urea 308.0
52 [3-(3-Phenyl-pyrido[2,3-b]pyrazin-6-yl)-ureido]-acetic acid-ethyl-ester 352.0
53 1-(3-Chloro-pyrido[2,3-b]pyrazin-6-yl)-3-ethyl-urea 252.1
54 1-Ethyl-3-[3-(4-fluoro-phenyl)-pyrido[2,3-b]pyrazin-6-yl]-urea 312.2
55 1-[3-(3-Chloro-4-fluoro-phenyl)-pyrido[2,3-b]pyrazin-6-yl]-3-ethyl- 346.2
urea
56 4-[6-(3-Ethyl-ureido)-pyrido[2,3-b]pyrazin-3-yl]-benzoic acid 338.1
57 N-{4-[6-(3-Ethyl-ureido)-pyrido[2,3-b]pyrazin-3-yl]-phenyl}-acetamide 350.9
58 1-[3-(2,4-Difluoro-phenyl)-pyrido[2,3-b]pyrazin-6-yl]-3-ethyl-urea 329.9
59 1-Ethyl-3-(3-morpholin-4-yl-pyrido[2,3-b]pyrazin-6-yl)-urea 303.1
60 1-Ethyl-3-[3-(4-methyl-piperazin-1-yl)-pyrido[2,3-b]pyrazin-6-yl]-urea 316.3
61 1-Ethyl-3-[3-(2-hydroxy-phenyl)-pyrido[2,3-b]pyrazin-6-yl]-urea 310.2
62 1-Ethyl-3-[3-(2-methoxy-ethylamino)-pyrido[2,3-b]pyrazin-6-yl]-urea 291.2
63 1-[3-(4-Chloro-3-trifluoromethyl-phenyl)-pyrido[2,3-b]pyrazin-6-yl]-3- 294.9
ethyl-urea
64 1-Ethyl-3-(3-phenoxy-pyrido[2,3-b]pyrazin-6-yl)-urea 310.2
65 1-[3-(Cyclopropylmethyl-amino)-pyrido[2,3-b]pyrazin-6-yl]-3-ethyl- 287.2
urea
66 1-Ethyl-3-{3-[(pyridin-4-ylmethyl)-amino]-pyrido[2,3-b]pyrazin-6-yl}- 324.2
urea
67 1-Ethyl-3-[3-(4-fluoro-benzyloxy)-pyrido[2,3-b]pyrazin-6-yl]-urea 341.9
68 1-Ethyl-3-(3-pyridin-4-yl-pyrido[2,3-b]pyrazin-6-yl)-urea 295.3
69 1-Ethyl-3-[3-(pyridin-3-yloxy)-pyrido[2,3-b]pyrazin-6-yl]-urea 311.3
70 1-Ethyl-3-[3-(tetrahydro-furan-2-ylmethoxy)-pyrido[2,3-b]pyrazin-6- 318.2
yl]-urea
71 1-Ethyl-3-[3-(4-morpholin-4-yl-phenyl)-pyrido[2,3-b]pyrazin-6-yl]- 379.4
urea
72 1-Ethyl-3-(3-hydroxy-pyrido[2,3-b]pyrazin-6-yl)-urea 234.3
73 1-Ethyl-3-[3-(3-methoxy-phenylsulfanyl)-pyrido[2,3-b]pyrazin-6-yl]- 355.9
urea
74 1-Ethyl-3-(3-quinolin-3-yl-pyrido[2,3-b]pyrazin-6-yl)-urea 345.1
75 1-(3-Benzo[b]thiophen-3-yl-pyrido[2,3-b]pyrazin-6-yl)-3-ethyl-urea 350.1
76 1-Ethyl-3-[3-(pyridin-2-ylsulfanyl)-pyrido[2,3-b]pyrazin-6-yl]-urea 327.3
77 1-[3-(4-Dimethylamino-phenyl)-pyrido[2,3-b]pyrazin-6-yl]-3-ethyl- 337.2
urea
78 N-{4-[6-(3-Ethyl-ureido)-pyrido[2,3-b]pyrazin-3-yl]-phenyl}- 387.3
methansulfonamide
79 1-Ethyl-3-[3-(1H-pyrazol-4-yl)-pyrido[2,3-b]pyrazin-6-yl]-urea 284.2
80 1-(3-Benzylsulfanyl-pyrido[2,3-b]pyrazin-6-yl)-3-ethyl-urea 340.0
81 1-Ethyl-3-[3-(4-methyl-[1,4]diazepan-1-yl)-pyrido[2,3-b]pyrazin-6-yl]- 330.5
urea
82 1-[3-(4-Amino-phenyl)-pyrido[2,3-b]pyrazin-6-yl]-3-ethyl-urea 309.4
83 1-(3-Amino-pyrido[2,3-b]pyrazin-6-yl)-3-ethyl-urea 233.3
84 1-Ethyl-3-pyrido[2,3-b]pyrazin-6-yl-urea 218.3
86 1-Ethyl-3-[3-(4-fluoro-2-hydroxy-phenyl)-pyrido[2,3-b]pyrazin-6-yl]- 328.2
urea
87 1-(3-Cyclopentyloxy-pyrido[2,3-b]pyrazin-6-yl)-3-ethyl-urea 302.3
88 1-Ethyl-3-[3-(4-hydroxy-piperidin-1-yl)-pyrido[2,3-b]pyrazin-6-yl]- 317.2
urea
89 (3-Phenyl-pyrido[2,3-b]pyrazin-6-yl)-urea 266.2
90 1-Ethyl-3-(3-pyrimidin-5-yl-pyrido[2,3-b]pyrazin-6-yl)-urea 296.3
91 1-Ethyl-3-(3-pyridin-3-yl-pyrido[2,3-b]pyrazin-6-yl)-urea 295.3
92 1-Allyl-3-(3-phenyl-pyrido[2,3-b]pyrazin-6-yl)-urea 306.3
93 1-Ethyl-3-(3-piperazin-1-yl-pyrido[2,3-b]pyrazin-6-yl)-urea 302.3
94 1-[3-(3-Chloro-pyridin-4-ylmethyl)-pyrido[2,3-b]pyrazin-6-yl]-3-ethyl- 329.3
urea
95 1-Ethyl-3-[3-(6-methoxy-pyridin-3-yl)-pyrido[2,3-b]pyrazin-6-yl]-urea 325.3
96 1-[3-(3,5-Dimethyl-isoxazol-4-yl)-pyrido[2,3-b]pyrazin-6-yl]-3-ethyl- 313.3
urea
97 1-Ethyl-3-[3-(4-trifluoromethoxy-phenyl)-pyrido[2,3-b]pyrazin-6-yl]- 378.3
urea
98 1-Ethyl-3-(3-furan-2-yl-pyrido[2,3-b]pyrazin-6-yl)-urea 284.2
99 1-Ethyl-3-[3-(2-methoxy-pyridin-3-yl)-pyrido[2,3-b]pyrazin-6-yl]-urea 325.3
100 1-[3-(2,4-Dimethoxy-phenyl)-pyrido[2,3-b]pyrazin-6-yl]-3-ethyl-urea 353.9
101 1-Ethyl-3-[3-(1H-pyrrol-2-yl)-pyrido[2,3-b]pyrazin-6-yl]-urea 383.3
102 1-Ethyl-3-[3-(6-morpholin-4-yl-pyridin-3-yl)-pyrido[2,3-b]pyrazin-6- 380.3
yl]-urea
103 1-Benzyl-3-ethyl-1-(3-phenyl-pyrido[2,3-b]pyrazin-6-yl)-urea 384.0
104 1-[3-(2-Amino-phenyl)-pyrido[2,3-b]pyrazin-6-yl]-3-ethyl-urea 309.2
105 1-Ethyl-3-[3-(4-hydroxymethyl-phenyl)-pyrido[2,3-b]pyrazin-6-yl]- 324.2
urea
106 1-[3-(3-Amino-phenyl)-pyrido[2,3-b]pyrazin-6-yl]-3-ethyl-urea 309.2
107 1-[3-(4-Acetyl-phenyl)-pyrido[2,3-b]pyrazin-6-yl]-3-ethyl-urea 336.1
108 1-[3-(2,3-Dihydro-benzofuran-5-yl)-pyrido[2,3-b]pyrazin-6-yl]-3-ethyl- 336.2
urea
109 1-[3-(4-Benzyloxy-3-fluoro-phenyl)-pyrido[2,3-b]pyrazin-6-yl]-3-ethyl- 418.3
urea
110 1-(2,3-Dihydroxy-propyl)-3-(3-phenyl-pyrido[2,3-b]pyrazin-6-yl)-urea 340.0
111 1-Ethyl-3-[3-(3-formyl-4-methoxy-phenyl)-pyrido[2,3-b]pyrazin-6-yl]- 352.1
urea
112 1-Ethyl-3-[3-(4-methansulfonyl-phenyl)-pyrido[2,3-b]pyrazin-6-yl]- 372.1
urea
113 N-{4-[6-(3-Ethyl-ureido)-pyrido[2,3-b]pyrazin-3-yl]-phenyl}- 409.3
succinamid acid
114 1-Ethyl-3-(3-pyridin-4-yl-pyrido[2,3-b]pyrazin-6-yl)-urea 295.3
Methansulfonic acid salt
115 1-[3-(2,6-Dimethoxy-pyridin-3-yl)-pyrido[2,3-b]pyrazin-6-yl]-3-ethyl- 354.9
urea
116 1-[3-(2,6-Dimethoxy-pyrimidin-4-yl)-pyrido[2,3-b]pyrazin-6-yl]-3- 356.1
ethyl-urea
117 1-[3-(2,4-Dioxo-1,2,3,4-tetrahydro-pyrimidin-5-yl)-pyrido[2,3- 328.4
b]pyrazin-6-yl]-3-ethyl-urea
118 1-Ethyl-3-[3-(1H-indol-5-yl)-pyrido[2,3-b]pyrazin-6-yl]-urea 333.0
119 1-(3-Chloroo-pyrido[2,3-b]pyrazin-6-yl)-3-ethyl-thiourea 268.0
120 1-Ethyl-3-{3-[4-(2-methoxy-ethoxy)-phenyl]-pyrido[2,3-b]pyrazin-6- 368.4
yl}-urea
121 Acrylic acid-4-[6-(3-ethyl-ureido)-pyrido[2,3-b]-pyrazin-3-yl]-phenyl- 364.1
ester
122 1-[3-(4-Cyano-phenyl)-pyrido[2,3-b]pyrazin-6-yl]-3-ethyl-urea 319.1
123 1-(3-Benzo[1,2,5]oxadiazol-4-yl-pyrido[2,3-b]pyrazin-6-yl)-3-ethyl- 336.0
urea
124 1-Ethyl-3-[3-(4-hydroxy-3-methoxy-phenyl)-pyrido[2,3-b]pyrazin-6- 340.0
yl]-urea
125 1-[3-(2,6-Dimethoxy-phenyl)-pyrido[2,3-b]pyrazin-6-yl]-3-ethyl-urea 354.2
126 1-[3-(3-Acetyl-phenyl)-pyrido[2,3-b]pyrazin-6-yl]-3-ethyl-urea 336.2
127 1-Ethyl-3-[3-(3-morpholin-4-yl-phenyl)-pyrido[2,3-b]pyrazin-6-yl]- 379.2
urea
128 1-[3-(6-Amino-pyridin-3-yl)-pyrido[2,3-b]pyrazin-6-yl]-3-ethyl-urea 310.2
129 3-{4-[6-(3-Ethyl-ureido)-pyrido[2,3-b]-pyrazin-3-yl]-phenoxy}- 382.3
propionic acid
130 1-Isopropyl-3-(3-phenyl-pyrido[2,3-b]pyrazin-6-yl)-urea 308.2
131 1-Cyclopentyl-3-(3-phenyl-pyrido[2,3-b]pyrazin-6-yl)-urea 333.9
132 1-Pentyl-3-(3-phenyl-pyrido[2,3-b]pyrazin-6-yl)-urea 336.2
133 N-{4-[6-(3-Ethyl-ureido)-pyrido[2,3-b]pyrazin-3-yl]-phenyl}- 363.2
acrylamide
134 1-tert-Butyl-3-(3-phenyl-pyrido[2,3-b]pyrazin-6-yl)-urea 322.1
135 1-(2-Hydroxy-ethyl)-3-(3-phenyl-pyrido[2,3-b]pyrazin-6-yl)-urea 310.2
136 1-Cyclobutyl-3-(3-phenyl-pyrido[2,3-b]pyrazin-6-yl)-urea 319.8
137 1-Allyl-3-[3-(4-nitro-phenyl)-pyrido[2,3-b]pyrazin-7-yl]-thiourea 367.1
138 1-Ethyl-1-(Ethylcarbamoyl)-3-(3-phenyl-pyrido[2,3-b]pyrazin-6-yl)- 365.1
urea
139 [3-(4-Hydroxy-phenyl)-pyrido[2,3-b]pyrazin-6-yl]-carbamic acid-allyl- 323.0
ester
140 (3-Phenyl-pyrido[2,3-b]pyrazin-6-yl)-carbamic acid-ethyl-ester 295.3
141 1-Ethyl-3-[3-(4-phenyl-piperazin-1-yl)-pyrido[2,3-b]pyrazin-6-yl]-urea 378.3
142 1-(2,2-Dimethyl-[1,3]dioxolan-4-ylmethyl)-3-(3-phenyl-pyrido[2,3- 380.2
b]pyrazin-6-yl)-urea
143 1,3-Bis-(3-phenyl-pyrido[2,3-b]pyrazin-6-yl)-urea 471.3
144 N-(3-Phenyl-pyrido[2,3-b]pyrazin-6-yl)-acetamidine 264.8
145 1-Ethyl-3-[3-(2-methoxy-pyridin-3-yl)-pyrido[2,3-b]pyrazin-6-yl]- 341.2
thiourea
146 1-(4-Hydroxy-butyl)-3-(3-phenyl-pyrido[2,3-b]pyrazin-6-yl)-urea 338.2
147 1-(3-Hydroxy-propyl)-3-(3-phenyl-pyrido[2,3-b]pyrazin-6-yl)-urea 324.2
148 1-Ethyl-3-{3-[4-(3-morpholin-4-yl-propoxy)-phenyl]-pyrido[2,3- 437.3
b]pyrazin-6-yl}-urea
149 1-Ethyl-3-[3-(4-hydroxy-phenyl)-pyrido[2,3-b]pyrazin-6-yl]-thiourea 326.1
150 1-Ethyl-3-(3-p-tolylamino-pyrido[2,3-b]pyrazin-6-yl)-urea 323.2
151 1-Ethyl-3-[3-(3-hydroxy-phenyl)-pyrido[2,3-b]pyrazin-6-yl]-urea 310.2
152 3-[6-(3-Ethyl-ureido)-pyrido[2,3-b]pyrazin-3-yl]-benzoic acid 338.2
153 1-[3-(3,4-Dimethoxy-phenyl)-pyrido[2,3-b]pyrazin-6-yl]-3-ethyl-urea 354.1
154 1-[3-(4-Amino-3-methoxy-phenyl)-pyrido[2,3-b]pyrazin-6-yl]-3-ethyl- 339.1
urea
155 3-[6-(3-Ethyl-ureido)-pyrido[2,3-b]-pyrazin-3-yl]-benzamide 337.1
156 1-[3-(3-Chloro-4-hydroxy-5-methoxy-phenyl)-pyrido[2,3-b]pyrazin-6- 374.2
yl]-3-ethyl-urea
157 1-Ethyl-3-(3-m-tolylamino-pyrido-[2,3-b]pyrazin-6-yl)-urea 323.3
158 1-Ethyl-3-[3-(4-methoxy-phenylamino)-pyrido[2,3-b]pyrazin-6-yl]-urea 339.0
159 1-[3-(4-Chloroo-phenylamino)-pyrido-[2,3-b]pyrazin-6-yl]-3-ethyl-urea 343.1
160 1-Ethyl-3-(3-o-tolylamino-pyrido[2,3-b]-pyrazin-6-yl)-urea 323.2
161 1-Ethyl-3-[3-(pyridin-3-ylamino)-pyrido-[2,3-b]pyrazin-6-yl]-urea 310.2
162 1-Ethyl-3-[3-(4-ethyl-phenylamino)-pyrido[2,3-b]pyrazin-6-yl]-urea 337.2
163 1-Ethyl-3-[3-(3-methoxy-4-methyl-phenylamino)-pyrido[2,3-b]pyrazin- 353.3
6-yl]-urea
164 1-Ethyl-3-[3-(4-hydroxy-phenylamino)-pyrido[2,3-b]pyrazin-6-yl]-urea 325.3
165 1-Ethyl-3-[3-(5-methyl-pyridin-2-yl-amino)-pyrido[2,3-b]pyrazin-6-yl]- 324.3
urea
166 1-Ethyl-3-[3-(1-methyl-1H-pyrazol-3-ylamino)-pyrido[2,3-b]pyrazin-6- 313.4
yl]-urea
167 1-Ethyl-3-[3-(4-fluoro-phenylamino)-pyrido[2,3-b]pyrazin-6-yl]-urea 327.4
168 1-(4-Hydroxy-butyl)-3-[3-(4-hydroxy-3-methoxy-phenyl)-pyrido[2,3- 384.3
b]pyrazin-6-yl]-urea
169 Phosphoric acid-mono-{4-[6-(3-ethyl-ureido)-pyrido-[2,3-b]pyrazin-3- 420.2
yl]-2-methoxy-phenyl}-ester
170 1-[3-(2-Chloro-4-hydroxy-phenyl)-pyrido[2,3-b]pyrazin-6-yl]-3-ethyl- 344.3
urea
171 1-[3-(4-Chloro-2-methoxy-phenyl)-pyrido[2,3-b]pyrazin-6-yl]-3-ethyl- 358.1
urea
172 1-[3-(3,5-Dimethyl-1H-pyrazol-4-yl)-pyrido[2,3-b]pyrazin-6-yl]-3- 312.2
ethyl-urea
173 1-Ethyl-3-[3-(1-methyl-1H-pyrazol-4-yl)-pyrido[2,3-b]pyrazin-6-yl]- 298.4
urea
174 1-[3-(5-Cyano-thiophen-2-yl)-pyrido[2,3-b]pyrazin-6-yl]-3-ethyl-urea 325.3
176 1-[3-(4-Hydroxy-3-methoxy-phenyl)-pyrido[2,3-b]pyrazin-6-yl]-3-(4- 453.4
morpholin-4-yl-butyl)-urea
177 1-[3-(3-Chloro-4-methoxy-phenylamino)-pyrido[2,3-b]pyrazin-6-yl]-3- 373.4
ethyl-urea
178 1-Ethyl-3-[3-(naphthalin-2-ylamino)-pyrido[2,3-b]pyrazin-6-yl]-urea 359.3
179 1-Ethyl-3-[3-(chinolin-3-ylamino)-pyrido[2,3-b]pyrazin-6-yl]-urea 360.3
180 1-[3-(3,5-Dimethoxy-phenylamino)-pyrido[2,3-b]pyrazin-6-yl]-3-ethyl- 369.3
urea
181 1-Ethyl-3-[3-(pyrazin-2-ylamino)-pyrido[2,3-b]pyrazin-6-yl]-urea 311.3
182 1-Ethyl-3-[3-(3-isopropoxy-phenylamino)-pyrido[2,3-b]pyrazin-6-yl]- 367.2
urea
183 1-Ethyl-3-[3-(4-hydroxy-3-methoxy-phenyl)-pyrido[2,3-b]pyrazin-6- 340.3
yl]-urea p-Toluolsulfonate (free base)
184 1-[3-(2-Chloro-pyridin-4-ylamino)-pyrido[2,3-b]pyrazin-6-yl]-3-ethyl- 344.3
urea
185 1-[3-(3,5-Dichloro-4-hydroxy-phenylamino)-pyrido[2,3-b]pyrazin-6- 393.2
yl]-3-ethyl-urea
186 1-[3-(3,5-Dichloro-4-hydroxy-phenyl)-pyrido[2,3-b]pyrazin-6-yl]-3- 369.2
ethyl-urea
187 1-[3-(3,4-Dimethoxy-phenylamino)-pyrido[2,3-b]pyrazin-6-yl]-3-ethyl- 369.2
urea
188 1-Ethyl-3-[3-(3-hydroxy-4-methyl-phenylamino)-pyrido[2,3-b]pyrazin- 339.1
6-yl]-urea
189 3-[6-(3-Ethyl-ureido)-pyrido[2,3-b]-pyrazin-3-ylamino]-5- 421.3
trifluoromethyl-benzoic acid
190 1-Ethyl-3-[3-(6-methoxy-pyridin-3-ylamino)-pyrido[2,3-b]pyrazin-6- 340.3
yl]-urea
191 1-[3-(3,5-Dimethyl-phenylamino)-pyrido[2,3-b]pyrazin-6-yl]-3-ethyl- 337.3
urea
192 1-[3-(4-Cyano-phenylamino)-pyrido-[2,3-b]pyrazin-6-yl]-3-ethyl-urea 334.1
194 1-Ethyl-3-[3-(4-hydroxy-3-methoxy-phenyl)-pyrido[2,3-b]pyrazin-6- 340.1
yl]-urea Hydrochloride (free base)
195 1-Ethyl-3-(3-phenyl-pyrido[2,3-b]pyrazin-6-yl)-thiourea 310.2
196 1-(3-Chloro-pyrido[2,3-b]pyrazin-6-yl)-3-cyclohexyl-urea 306.2
197 1-Ethyl-3-[3-(4-hydroxy-3-methoxy-phenylamino)-pyrido[2,3- 355.3
b]pyrazin-6-yl]-urea
198 1-Ethyl-3-[3-(4-hydroxy-3,5-dimethyl-phenyl)-pyrido[2,3-b]pyrazin-6- 338.3
yl]-urea
199 3-Ethyl-1-phenethyl-1-(3-phenyl-pyrido[2,3-b]pyrazin-6-yl)-urea 398.2
200 1-Allyl-3-{3-[4-(tetrahydro-pyran-2-yloxy)-phenyl]-pyrido[2,3- 422.2
b]pyrazin-6-yl}-thiourea
201 3-Ethyl-1-[3-(4-methoxy-phenyl)-pyrido[2,3-b]pyrazin-6-yl]-1-propyl- 366.3
urea
202 3-Ethyl-1-(3-phenyl-pyrido[2,3-b]pyrazin-6-yl)-1-(2-piperidin-1-yl- 405.4
ethyl)-urea Hydrochloride (free base)
203 N-{4-[6-(3-Ethyl-ureido)-pyrido[2,3-b]-pyrazin-3-yl]-phenyl}-2- 427.3
phenyl-acetamide
204 1-(4-Hydroxy-butyl)-3-(3-phenyl-pyrido[2,3-b]pyrazin-6-yl)-urea 338.3
Hydrochloride (free base)
205 Acetic acid-4-[3-(3-phenyl-pyrido[2,3-b]pyrazin-6-yl)-ureido]-butyl 380.4
ester
206 1-(4-Amino-butyl)-3-(3-phenyl-pyrido[2,3-b]pyrazin-6-yl)-urea 337.3
207 1-(5-Hydroxy-pentyl)-3-(3-phenyl-pyrido[2,3-b]pyrazin-6-yl)-urea 352.3
208 3-[6-(3-Ethyl-ureido)-pyrido[2,3-b]-pyrazin-3-yl]-N-methyl-benzamide 351.2
209 1-Ethyl-3-[3-(2-methoxy-5-methyl-phenyl)-pyrido[2,3-b]pyrazin-6-yl]- 338.1
urea
210 1-Ethyl-3-(3-p-tolyl-pyrido[2,3-b] pyrazin-6-yl)-urea 308.2
211 1-Ethyl-3-[3-(methyl-p-tolyl-amino)-pyrido[2,3-b]pyrazin-6-yl]-urea 337.1
212 1-Ethyl-3-[3-(2-p-tolyl-ethylamino)-pyrido[2,3-b]pyrazin-6-yl]-urea 351.1
213 1-Ethyl-3-[3-(4-methyl-benzylamino)-pyrido[2,3-b]pyrazin-6-yl]-urea 337.2
214 1-Ethyl-3-[3-(3-fluoro-4-methyl-phenyl-amino)-pyrido[2,3-b]pyrazin-6- 341.2
yl]-urea
215 1-[3-(3,4-Dimethyl-phenylamino)-pyrido[2,3-b]pyrazin-6-yl]-3-ethyl- 337.1
urea
216 1-Ethyl-3-[3-(4-isopropyl-phenylamino)-pyrido[2,3-b]pyrazin-6-yl]- 351.1
urea
217 1-(4-Morpholin-4-yl-butyl)-3-(3-phenyl-pyrido[2,3-b]pyrazin-6-yl)- 407.3
urea
218 N-{4-[3-(3-Phenyl-pyrido[2,3-b]pyrazin-6-yl)-ureido]-butyl}-acetamide 379.3
219 1-[3-(3-Amino-4-methyl-phenyl)-pyrido[2,3-b]pyrazin-6-yl]-3-ethyl- 323.3
urea
220 1-[3-(3-Acetyl-2-fluoro-phenyl)-pyrido[2,3-b]pyrazin-6-yl]-3-ethyl- 354.2
urea
221 1-Ethyl-3-[3-(4-methoxy-3-methyl-phenyl)-pyrido[2,3-b]pyrazin-6-yl]- 338.3
urea
222 1-[3-(6-Ethoxy-pyridin-3-yl)-pyrido[2,3-b]pyrazin-6-yl]-3-ethyl-urea 339.3
223 1-Ethyl-3-[3-(2-fluoro-4-methyl-phenyl)-pyrido[2,3-b]pyrazin-6-yl]- 326.2
urea
224 1-Ethyl-3-[3-(3-fluoro-4-methoxy-phenyl)-pyrido[2,3-b]pyrazin-6-yl]- 342.1
urea
225 1-Ethyl-3-[3-(2-fluoro-3-methoxy-phenyl)-pyrido[2,3-b]pyrazin-6-yl]- 342.1
urea
226 1-Ethyl-3-[3-(3,4,5-trimethoxy-phenyl)-pyrido[2,3-b]pyrazin-6-yl]-urea 384.2
227 1-[3-(3,5-Difluoro-2-methoxy-phenyl)-pyrido[2,3-b]pyrazin-6-yl]-3- 360.3
ethyl-urea
228 1-Ethyl-3-[3-(4-trifluoromethyl-phenyl)-pyrido[2,3-b]pyrazin-6-yl]- 362.3
urea
229 1-Ethyl-3-[3-(2,3,4-trimethoxy-phenyl)-pyrido[2,3-b]pyrazin-6-yl]-urea 384.3
230 1-[3-(3-Chloro-4-methoxy-phenyl)-pyrido[2,3-b]pyrazin-6-yl]-3-ethyl- 358.2
urea
231 1-Ethyl-3-[3-(3-fluoro-4-hydroxy-phenyl)-pyrido[2,3-b]pyrazin-6-yl]- 328.3
urea
232 1-Ethyl-3-[3-(6-fluoro-pyridin-3-yl)-pyrido[2,3-b]pyrazin-6-yl]-urea 313.3
233 1-[3-(2,4-Dimethyl-thiazol-5-yl)-pyrido-[2,3-b]pyrazin-6-yl]-3-ethyl- 329.4
urea
234 1-Ethyl-3-[3-(2-methoxy-phenyl)-pyrido[2,3-b]pyrazin-6-yl]-urea 324.3
235 1-[3-(2-Chloro-pyridin-4-yl)-pyrido[2,3-b]pyrazin-6-yl]-3-ethyl-urea 329.3
236 1-[3-(5-Acetyl-thiophen-2-yl)-pyrido[2,3-b]pyrazin-6-yl]-3-ethyl-urea 342.3
237 1-[3-(5-Chloro-thiophen-2-yl)-pyrido[2,3-b]pyrazin-6-yl]-3-ethyl-urea 334.1
238 1-Ethyl-3-[3-(3-methoxy-phenyl)-pyrido[2,3-b]pyrazin-6-yl]-urea 324.1
239 1-[3-(3-Bromo-5-methoxy-phenyl)-pyrido[2,3-b]pyrazin-6-yl]-3-ethyl- 402.3
urea
240 1-[3-(Benzothiazol-6-ylamino)-pyrido[2,3-b]pyrazin-6-yl]-3-ethyl-urea 366.3
241 1-Ethyl-3-[3-(4-methyl-3-trifluoroomethyl-phenylamino)-pyrido[2,3- 391.4
b]pyrazin-6-yl]-urea
242 1-[3-(3-Cyano-4-methyl-phenylamino)-pyrido[2,3-b]pyrazin-6-yl]-3- 348.3
ethyl-urea
243 1-Ethyl-3-[3-(4-phenoxy-phenylamino)-pyrido[2,3-b]pyrazin-6-yl]-urea 401.3
244 1-[3-(4-Chloro-3-methyl-phenylamino)-pyrido[2,3-b]pyrazin-6-yl]-3- 357.3
ethyl-urea
245 1-[3-(2-Chloro-4-methyl-phenylamino)-pyrido[2,3-b]pyrazin-6-yl]-3- 357.1
ethyl-urea
246 1-Ethyl-3-[3-(3-trifluoroomethyl-phenylamino)-pyrido[2,3-b]pyrazin-6- 377.4
yl]-urea
247 1-[3-(2-Chloro-4-trifluoromethoxy-phenylamino)-pyrido[2,3-b]pyrazin- 427.3
6-yl]-3-ethyl-urea
248 1-[3-(4-Chloro-2-methoxy-5-methyl-phenylamino)-pyrido[2,3- 387.4
b]pyrazin-6-yl]-3-ethyl-urea
249 1-Ethyl-3-[3-(4-methylsulfanyl-phenyl)-pyrido[2,3-b]pyrazin-6-yl]-urea 340.3
250 3-[6-(3-Ethyl-ureido)-pyrido[2,3-b]pyrazin-3-yl]-benzolsulfonamide 373.3
251 N-{4-[3-(3-Phenyl-pyrido[2,3-b]pyrazin-6-yl)-ureido]-butyl}- 415.3
methansulfonamide

II.2 NMR Spectroscopic Data and Melting Points for Selected Compounds
Compound 1: 1-allyl-3-(3-phenyl-pyrido[2,3-b]pyrazin-6-yl)-thiourea
Melting point: 239-240° C. (Decomp.)
1H-NMR (d6-DMSO): δ=4.40 (m, 2H), 5.30 (d, 1H), 5.60 (d, 1H), 6.07-6.17 (m, 1H), 7.55-7.70 (m, 4H), 8.35 (d, 2H), 8.45 (d, 1H), 9.50 (s, 1H), 11.35 (s, 1H), 12.55 (m, 1H).
Compound 2: 1-(2-methyl-allyl)-3-(3-naphth-2-yl-pyrido[2,3-b]pyrazin-6-yl)-thiohurea
Mp.: 239-240° C. (Decomp.)
1H-NMR (d6-DMSO): δ=1.94 (s, 3H), 4.32 (m, 2H), 5.07 (s, 1H), 5.28 (s, 1H), 7.60-7.69 (m, 3H), 8.00-8.05 (m, 1H), 8.07-8.12 (m, 1H), 8.14 (d, 1H), 8.42-8.51 (m, 2H), 8.98 (s, 1H), 9.68 (s, 1H), 11.32 (s, 1H), 12.78 (m, 1H).
Compound 3: 1-[3-(4-methoxy-phenyl)-pyrido[2,3-b]pyrazin-6-yl]-3-(2-methyl-allyl)-thiourea
Mp.: 251-252° C. (Decomp.)
1H-NMR (d6-DMSO): δ=1.92 (s, 3H), 3.85 (s, 3H), 4.27-4.35 (m, 2H), 5.02 (s, 1H), 5.24 (s, 1H), 7.15 (d, 2H), 7.58 (d, 1H), 8.31 (d, 2H), 8.41 (d, 1H), 9.46 (s, 1H), 11.29 (s, 1H), 12.68 (m, 1H).
Compound 4: 1-(2-methyl-allyl)-3-(3-phenyl-pyrido[2,3-b]pyrazin-6-yl)-thiourea
Mp.: 225-226° C. (Decomp.)
1H-NMR (d6-DMSO): δ=1.90 (s, 3H), 4.30-4.35 (m, 2H), 5.01 (s, 1H), 5.22 (s, 1H), 7.55-7.80 (m, 4H), 8.30-8.38 (m, 2H), 8.45 (d, 1H), 9.52 (s, 1H), 11.32 (s, 1H), 12.65 (m, 1H).
Compound 5: 1-allyl-3-(3-naphth-2-yl-pyrido[2,3-b]pyrazin-6-yl)-thiourea
Mp.: 242-243° C. (Decomp.)
1H-NMR (d6-DMSO): δ=4.42 (m, 2H), 5.37 (d, 1H), 5.65 (d, 1H), 6.07-6.19 (m, 1H), 7.57-7.68 (m, 3H), 7.97-8.05 (m, 1H), 8.07-8.19 (m, 2H), 8.40-8.52 (m, 2H), 8.99 (s, 1H), 9.70 (s, 1H), 11.36 (s, 1H), 12.56 (t, 1H).
Compound 6: 1-allyl-3-[3-(4-methoxy-phenyl)-pyrido[2,3-b]pyrazin-6-yl]-thiourea
Mp.: 240-241° C. (Decomp.)
1H-NMR (d6-DMSO): δ=3.87 (s, 3H), 4.36-4.42 (m, 2H), 5.32 (d, 1H), 5.60 (d, 1H), 6.06-6.16 (m, 1H), 7.16 (d, 2H), 7.60 (d, 1H), 8.32 (d, 2H), 8.42 (d, 1H), 9.56 (s, 1H), 11.29 (s, 1H), 12.56 (m, 1H).
Compound 7: 1-allyl-3-[3-(4-hydroxy-phenyl)-pyrido[2,3-b]pyrazin-6-yl]-thiourea-hydrochloride
Mp.: 160-161° C. (Decomp.)
1H-NMR (d6-DMSO): δ=4.36-4.43 (m, 2H), 5.31 (d, 1H), 5.59 (d, 1H), 6.05-6.16 (m, 1H), 6.97 (d, 2H), 7.57 (d, 1H), 8.20 (d, 2H), 8.40 (d, 1H), 9.41 (s, 1H), 10.17 (bs, 1H), 11.24 (s, 1H), 12.56 (m, 1H).
Compound 8: 1-(3-naphth-2-yl-pyrido[2,3-b]pyrazin-6-yl)-3-(4-nitro-phenyl)-thiourea
Mp.: 260-261° C. (Decomp.)
1H-NMR (d6-DMSO): δ=7.61-7.68 (m, 3H), 7.72 (d, 2H), 7.75 (d, 1H), 8.01-8.06 (m, 1H), 8.16 (m, 2H), 8.26 (d, 2H), 8.53 (d, 1H), 8.58 (d, 1H), 9.04 (s, 1H), 9.62 (s, 1H), 9.76 (s, 1H), 11.81 (s, 1H).
Compound 9: 1-[3-(4-methoxy-phenyl)-pyrido[2,3-b]pyrazin-6-yl]-3-(4-nitro-phenyl)-thiourea
Mp.: 250-251° C. (Decomp.)
1H-NMR (d6-DMSO): δ=3.85 (s, 3H), 7.17 (d, 2H), 7.71 (d, 2H), 8.21 (d, 2H), 8.22-8.27 (m, 1H), 8.36-8.42 (m, 3H), 9.53 (s, 1H), 9.65 (s, 1H), 11.77 (s, 1H).
Compound 10: 1-tert.butyl-3-(3-phenyl-pyrido[2,3-b]pyrazin-6-yl)-thiourea
Mp.: 227° C. (Decomp.)
1H-NMR (d6-DMSO): δ=1.65 (s, 9H), 7.53-7.69 (m, 4H), 8.34 (d, 2H), 8.41 (d, 1H), 9.51 (s, 1H), 10.98 (s, 1H), 12.75 (s, 1H).
Compound 11: 1-cyclopropyl-3-(3-phenyl-pyrido[2,3-b]pyrazin-6-yl)-thiourea
Mp.: 233-234° C.
1H-NMR (d6-DMSO): δ=0.70-0.80 (m, 2H), 0.91-1.00 (m, 2H), 3.20-3.28 (m, 1H), 7.51-7.72 (m, 4H), 8.36 (d, 2H), 8.45 (d, 1H), 9.52 (s, 1H), 11.31 (s, 1H), 12.45 (s, 1H).
Compound 12: 1-methyl-3-(3-phenyl-pyrido[2,3-b]pyrazin-6-yl)-thiourea
Mp.: 253-254° C.
1H-NMR (d6-DMSO): δ=3.25 (s, 3H), 7.59-7.67 (m, 4H), 8.38 (d, 2H), 8.46 (d, 1H), 9.52 (s, 1H), 11.31 (s, 1H), 12.10 (s, 1H).
Compound 13: 1-benzyl-3-(3-phenyl-pyrido[2,3-b]pyrazin-6-yl)-thiourea
Mp.: 232-233° C.
1H-NMR (d6-DMSO): δ=4.96 (m, 2H), 7.37-7.48 (m, 3H), 7.54-7.67 (m, 6H), 8.32 (d, 2H), 8.47 (d, 1H), 9.52 (s, 1H), 11.43 (s, 1H), 12.91 (s, 1H).
Compound 14: 1-(4-fluoro-phenyl)-3-(3-phenyl-pyrido[2,3-b]pyrazin-6-yl)-thiourea
Mp.: 225-226° C.
1H-NMR (d6-DMSO): δ=7.33 (m, 2H), 7.57-7.65 (m, 3H), 7.70-7.81 (m, 3H), 8.34 (d, 2H), 8.54 (d, 1H), 9.57 (s, 1H), 11.62 (s, 1H).
Compound 15: 1-(3-phenyl-pyrido[2,3-b]pyrazin-6-yl)-3-p-tolyl-urea
Mp.: 298-299° C.
1H-NMR (d6-DMSO): δ=2.29 (s, 3H), 7.20 (d, 2H), 7.52 (d, 2H), 7.59-7.67 (m, 3H), 7.80 (d, 1H), 8.38 (d, 2H), 8.44 (d, 1H), 9.59 (s, 1H), 10.36 (s, 1H), 11.46 (s, 1H).
Compound 16: 1-(4-chloro-3-trifluoromethyl-phenyl)-3-(3-phenyl-pyrido[2,3-b]pyrazin-6-yl)-urea
Mp.: 250° C.
1H-NMR (d6-DMSO): δ=7.58-7.67 (m, 3H), 7.74 (d, 1H), 7.80 (d, 1H), 7.87 (d, 1H), 8.21 (s, 1H), 8.39 (d, 2H), 8.48 (d, 1H), 9.53 (s, 1H), 10.55 (s, 1H), 11.82 (s, 1H).
Compound 17: 1-(2-morpholin-4-yl-ethyl)-3-(3-phenyl-pyrido[2,3-b]pyrazin-6-yl)-urea
Mp.: 226° C.
1H-NMR (d6-DMSO): δ=2.45-2.67 (m, 6H), 3.40-3.48 (m, 2H), 3.60-3.69 (m, 4H), 7.55-7.70 (m, 4H), 8.30-8.40 (m, 3H), 9.29 (s, 1H), 9.42 (s, 1H), 10.18 (s, 1H).
Compound 18: 1-cyclohexyl-3-(3-phenyl-pyrido[2,3-b]pyrazin-6-yl)-thiourea
Mp.: 230-232° C.
1H-NMR (d6-DMSO): δ=1.50-1.75 (m, 6H), 1.80-2.00 (m, 4H), 7.55-7.70 (m, 4H), 8.37 (d, 2H), 8.45 (d, 1H), 9.55 (s, 1H), 11.20 (s, 1H), 12.80 (s, 1H).
Compound 19: 1-isopropyl-3-(3-phenyl-pyrido[2,3-b]pyrazin-6-yl)-thiourea
Mp.: 229-230° C.
1H-NMR (d6-DMSO): δ=1.40 (d, 6H), 4.40-4.50 (m, 1H), 7.58-7.66 (m, 4H), 8.36 (d, 2H), 8.44 (d, 1H), 9.52 (s, 1H), 11.20 (s, 1H), 12.48 (s, 1H).
Compound 20: 1-furan-2-ylmethyl-3-(3-phenyl-pyrido[2,3-b]pyrazin-6-yl)-thiourea
Mp.: 250° C. (Decomp.)
1H-NMR (d6-DMSO): δ=4.95 (s, 2H), 6.55 (m, 1H), 6.68 (d, 1H), 7.59-7.68 (m, 4H), 7.74 (d, 1H), 8.37 (d, 2H), 8.48 (d, 1H), 9.55 (s, 1H), 11.45 (s, 1H), 12.83 (s, 1H).
Compound 21: 1-cyclopropyl-3-(3-phenyl-pyrido[2,3-b]pyrazin-6-yl)-urea
Mp.: 158-160° C.
1H-NMR (d6-DMSO): δ=0.52-0.60 (m, 2H), 0.72-0.82 (m, 2H), 2.70-2.79 (m, 1H), 7.57-7.65 (m, 3H), 7.71 (d, 1H), 8.34 (d, 2H), 8.38 (d, 1H), 9.21 (s, 1H), 9.46 (s, 1H), 10.12 (s, 1H).
Compound 22: 1-methyl-3-[3-(4-nitro-phenyl)-pyrido[2,3-b]pyrazin-6-yl]-thiourea
Mp.: 270° C.
1H-NMR (d6-DMSO): δ=3.25 (s, 3H), 7.70 (d, 1H), 8.44 (d, 2H), 8.50 (d, 1H), 8.64 (d, 2H), 9.64 (s, 1H), 11.38 (s, 1H), 12.03 (s, 1H).
Compound 23: 1-[3-(4-hydroxy-phenyl)-pyrido[2,3-]pyrazin-6-yl]-3-methyl-thiourea
Mp.: 282° C.
1H-NMR (d6-DMSO): δ=3.25 (s, 3H), 6.98 (d, 2H), 7.57 (d, 1H), 8.26 (d, 2H), 8.40 (d, 1H), 9.45 (s, 1H), 10.18 (s, 1H), 11.25 (s, 1H), 12.10 (s, 1H).
Compound 24: 1-allyl-3-[3-(4-nitro-phenyl)-pyrido[2,3-b]pyrazin-6-yl]-thiourea
Mp.: 244° C. (Decomp.)
1H-NMR (d6-DMSO): δ=4.40 (s, 2H), 5.36 (d, 1H), 5.59 (d, 1H), 6.08-6.15 (m, 1H), 7.71 (d, 1H), 8.46 (d, 2H), 8.51 (d, 1H), 8.60 (d, 2H), 9.64 (s, 1H), 11.45 (s, 1H), 12.51 (t, 1H).
Compound 25: 1-allyl-3-[3-(4-hydroxy-phenyl)-pyrido[2,3-b]pyrazin-6-yl]-urea
Mp.: 240° C. (Decomp.)
1H-NMR (d6-DMSO): δ=3.98 (s, 2H), 5.19 (d, 1H), 5.37 (d, 1H), 5.96-6.05 (m, 1H), 6.97 (d, 2H), 7.59 (d, 1H), 8.22 (d, 2H), 8.33 (d, 1H), 9.38 (s, 1H), 9.45 (s, 1H), 10.13 (s, 1H), 10.18 (s, 1H).
Compound 26: 4-[6-(3-allyl-thioureido)-pyrido[2,3-b]pyrazin-3-yl]-benzoic acid ethyl ester
Mp.: 223-224° C.
1H-NMR (d6-DMSO): δ=1.39 (t, 3H), 4.35-4.42 (m, 4H), 5.35 (d, 1H), 5.60 (d, 1H), 6.08-6.15 (m, 1H), 7.68 (d, 1H), 8.17 (d, 2H), 8.47 (d, 2H), 8.50 (d, 1H), 9.60 (s, 1H), 11.40 (s, 1H), 12.52 (t, 1H).
Compound 27: 1-allyl-3-[3-(3-hydroxy-phenyl)-pyrido[2,3-b]pyrazin-6-yl]-thiourea
Mp.: 205° C. (Decomp.)
1H-NMR (d6-DMSO): δ=4.41 (s, 2H), 5.33 (d, 1H), 5.58 (d, 1H), 6.07-6.15 (m, 1H), 6.99 (d, 1H), 7.42 (t, 1H), 7.64 (d, 1H), 7.72 (s, 1H), 7.77 (d, 1H), 8.46 (d, 1H), 9.45 (s, 1H), 9.80 (s, 1H), 11.37 (s, 1H), 12.55 (s, 1H).
Compound 28: 1-allyl-3-(3-benzo[1,3]dioxol-5-yl-pyrido[2,3-b]pyrazin-6-yl)-thiourea
Mp.: 218-220° C. (Decomp.)
1H-NMR (d6-DMSO): δ=4.40 (s, 2H), 5.31 (d, 1H), 5.60 (d, 1H), 6.08-6.20 (m, 3H), 7.16 (d, 1H), 7.61 (d, 1H), 7.90 (s, 1H), 7.96 (d, 1H), 8.43 (d, 1H), 9.49 (s, 1H), 11.34 (s, 1H), 12.58 (s, 1H).
Compound 29: 1-[3-(4-hydroxy-phenyl)-pyrido[2,3-b]pyrazin-6-yl]-3-prop-2-inyl-thiourea
Mp.: 350° C. (Decomp.)
1H-NMR (d6-DMSO): δ=2.09 (s, 1H), 2.44 (s, 2H), 6.99 (d, 2H), 7.19 (s, 1H), 7.44 (s, 1H), 8.24 (d, 2H), 8.26 (d, 1H), 9.29 (s, 1H), 10.08 (s, 1H), 11.81 (s, 1H).
Compound 30: 1-allyl-3-[3-(4-hydroxy-phenyl)-pyrido[2,3-b]pyrazin-6-yl]-thiourea
Mp.: 230° C. (Decomp.)
1H-NMR (d6-DMSO): δ=4.40 (s, 2H), 5.34 (d, 1H), 5.60 (d, 1H), 6.07-6.15 (m, 1H), 6.98 (d, 2H), 7.58 (d, 1H), 8.24 (d, 2H), 8.42 (d, 1H), 9.45 (s, 1H), 10.19 (s, 1H), 11.34 (s, 1H), 12.60 (s, 1H).
Compound 31: 1-[3-(4-hydroxy-phenyl)-pyrido[2,3-b]pyrazin-6-yl]-3-((E)-propenyl)-thiourea
1H-NMR (d6-DMSO): δ=2.12 (d, 3H), 5.17 (m, 1H), 6.96 (d, 2H), 7.22-7.26 (m, 1H), 7.59 (d, 1H), 8.25 (d, 2H), 8.45 (d, 1H), 9.48 (s, 1H), 10.20 (s, 1H), 11.56 (s, 1H), 14.67 (s, 1H).
Compound 32: 1-allyl-3-[2,3-bis-(4-hydroxy-phenyl)-pyrido[2,3-b]pyrazin-6-yl]-thiourea
Mp.: 270° C. (Decomp.)
1H-NMR (d6-DMSO): δ=4.40 (s, 2H), 5.25 (d, 1H), 5.50 (d, 1H), 6.02-6.13 (m, 1H), 6.74 (d, 2H), 6.76 (d, 2H), 7.31 (d, 2H), 7.36 (d, 2H), 7.62 (d, 1H), 8.42 (d, 1H), 9.78 (s, 1H), 9.85 (s, 1H), 11.30 (s, 1H), 12.47 (s, 1H).
Compound 33: 1-[2,3-bis-(4-hydroxy-phenyl)-pyrido[2,3-b]pyrazin-6-yl]-3-((E)-propenyl)-thiourea
Mp.: 240° C. (Decomp.)
1H-NMR (d6-DMSO): δ=2.05 (d, 3H), 5.10-5.18 (m, 1H), 6.74 (d, 2H), 6.76 (d, 2H), 7.20-7.26 (m, 1H), 7.34 (d, 2H), 7.39 (d, 2H), 7.63 (d, 1H), 8.45 (d, 1H), 9.79 (s, 1H), 9.89 (s, 1H), 11.55 (s, 1H), 14.56 (d, 1H).
Compound 34: 1-allyl-3-[2-(4-hydroxy-phenyl)-pyrido[2,3-b]pyrazin-6-yl]-thiourea
Mp.: 260° C. (Decomp.)
1H-NMR (d6-DMSO): δ=4.40 (s, 2H), 5.28 (d, 1H), 5.48 (d, 1H), 6.03-6.12 (m, 1H), 6.96 (d, 2H), 7.66 (d, 1H), 8.16 (d, 2H), 8.43 (d, 1H), 9.52 (s, 1H), 10.06 (s, 1H), 11.31 (s, 1H), 12.40 (s, 1H).
Compound 35: 1-phenethyl-3-(3-phenyl-pyrido[2,3-]pyrazin-6-yl)-urea
Mp.: 250° C. (Decomp.)
1H-NMR (d6-DMSO): δ=2.88-2.95 (m, 2H), 3.52-3.60 (m, 2H), 7.18 (t, 1H), 7.28 (t, 2H), 7.42 (d, 2H), 7.58-7.68 (m, 4H), 8.37 (d, 3H), 9.25 (s, 1H), 9.48 (s, 1H), 10.18 (s, 1H).
Compound 36: 1-(2,3-di-pyridin-2-yl-pyrido[2,3-b]pyrazin-6-yl)-3-ethyl-urea
Mp.: 236-237° C.
1H-NMR (d6-DMSO): δ=1.13-1.22 (m, 3H), 3.28-3.39 (m, 2H), 3.60-3.69 (m, 4H), 7.31-7.39 (m, 2H), 7.79 (d, 1H), 7.91-7.99 (m, 4H), 8.26 (d, 1H), 8.29 (d, 1H), 8.47 (d, 1H), 9.08 (s, 1H), 10.20 (s, 1H).
Compound 37: 1-(2,3-dimethyl-pyrido[2,3-b]pyrazin-6-yl)-3-ethyl-urea
Mp.: 246-248° C.
1H-NMR (d6-DMSO): δ=1.17 (t, 3H), 2.64 (s, 3H), 2.67 (s, 3H), 3.24-3.40 (m, 2H), 7.55 (d, 1H), 8.24 (d, 1H), 9.14 (s, 1H), 9.91 (s, 1H).
Compound 85: 1-ethyl-3-(3-imidazol-1-yl-pyrido[2,3-b]pyrazin-6-yl)-urea
1H-NMR (d6-DMSO): δ=1.20 (t, 3H), 3.26-3.32 (m, 2H), 7.26 (s, 1H), 7.75 (d, 1H), 8.20 (s, 1H), 8.40 (d, 1H), 8.79 (s, 1H), 8.84 (s, 1H), 9.39 (s, 1H), 10.16 (s, 1H) ppm.
Compound 137: 1-allyl-3-[3-(4-nitro-phenyl)-pyrido[2,3-b]pyrazin-7-yl]-thiourea
Mp.: 250° C. (Decomp.)
1H-NMR (d6-DMSO): δ=4.23 (s, 2H), 5.19 (d, 1H), 5.29 (d, 1H), 5.90-6.00 (m, 1H), 8.46 (d, 2H), 8.55 (s, 1H), 8.64 (d, 2H), 8.92 (s, 1H), 9.23 (s, 1H), 9.77 (s, 1H), 10.35 (s, 1H).
III. Evidence of the Kinase Inhibition of Compounds According to the Invention
III.1 Cell-Free Kinase Assays (Using ALPHA Technology)
The inhibitory effect of the compounds according to the invention was tested on various serine/threonine, tyrosine and lipid kinases in enzymatic assays. Recombinant human kinases such as, for example, Erk2, PI3Kalpha, -beta, -gamma, -delta, p38alpha, p38gamma, Jnk1, Jnk2 and others were used in this case, partly as full-length kinases, partly as shortened fragments, but at least consisting of the functional kinase domains. The commercial kinase proteins (Proqinase, Upstate) were used as recombinant fusion proteins with GST (glutathion-S-transferase) or His-Tag. Depending on the type of substrate, the various kinase reactions were quantified by means of suitable ALPHA™ beads (Perkin-Elmer).
Testing
The substance testing is described in detail hereinafter for the Erk assay. Selected test results of the Erk2, PD3Kalpha, p38alpha and Jnk2 assays are given below. To determine the IC50 value, the potential inhibitor substances were investigated in 10 semi-logarithmically graded concentrations of 3.16 nM-100 μM.
a) MAPK-ALPHAs (e.g. Erk2): the test substance, 0.625 ng Erk2 (#14-173, Upstate), 10 μM ATP and 15 nM biotinylated MBP (myelin basic protein) substrate were incubated on a 384-well Optiplate (Perkin-Elmer) in a volume of 15 μl for 1 h in 25 mM Tris, 10 mM MgCl2, 0.1% Tween-20, 100 μM NaVO4, 2 mM DTT at pH 7.5. The kinase reaction was then stopped by adding 10 μl of the ALPHA bead mixes (10 μg/ml, #6760617/Perkin-Elmer) pre-incubated with anti-phospho MBP antibody (320 pM, #05-429/Upstate) in 25 mM Tris, 200 mM NaCl, 110 nM EDTA and 0.3% BSA and left to stand overnight.
b) PI3K-ALPHAs (e.g. PI3Kalpha): the test substance, 1 ng PI3Kalpha (#14-602, Upstate), 100 μM ATP and 20 μM PIP2 substrate (#P4508, Echelon) were incubated on a 384-well Optiplate (Perkin-Elmer) for 1 h in 50 mM Hepes, 50 mM NaCl, 5 mM MgCl2, 0.05% Chaps, 5 mM DTT at pH 7.4. The kinase reaction was then stopped by adding ALPHA bead mixes (10 μg/ml, #6760603/Perkin-Elmer) pre-incubated with 1 nM GST:Grp1 fusion protein (Upstate) and 15 nM biotinylated PIP3 (#C-39B6/Echelon) in 50 mM Hepes, 50 mM NaCl, 50 mM EDTA and 0.1% BSA and left to stand overnight.
The fluorescence was detected the following morning in a Fusion™aα-system (Perkin-Elmer).
Evaluation
The %-inhibition values per substance concentration were calculated by means of the following formula from the raw data determined in the Fusion™
Figure US08937068-20150120-P00010
:
% Kinase inhibition ( Sample ) = 100 - ( 100 × Mean ( Sample ) - Mean ( 0 % Control ) Mean ( 100 % Control ) - Mean ( 0 % Control ) )
Eight determinations were made for each control and two for the substance samples. The 0% control either contains no ATP or no substrate, the 100% control (fully active kinase) contains no test substance. The IC50 values were determined using GraphPadPrism.
The compounds according to the invention showed effective inhibition of Erk, PI3K, p38alpha and Jnk1+Jnk2 with IC50 values of up to 6 nM (see Table 2).
TABLE 2
MAPK and PI3Kalpha kinase assay test results (IC50 [μM] bei
10 μM bzw. 100 μM* ATP)
Compound Erk2 PI3Kalpha p38alpha Jnk1 + Jnk2
1 1 0.9 >31.6 >31.6
25 0.076 1.5 >100 >31.6
30 0.392 0.37 >100 21.8
43 0.006 6.2 >100 5.5
44 2.5 3.8 >100 1.5
46 9.5 >100 0.495 >100
82 0.944 6.4 >31.6 1.1
124 0.062 0.49 >100 1.0
146 0.155 1.1 >100 3.3
147 0.711 4.1 >100 14.2
148 1.8 1.4 >100 13.7
149 0.954 3.5 >100 >31.6
150 18.7 0.129 18.1 >100
151 0.104 1.5 >100 2.9
152 1.9 5.3 >100 12.1
153 0.939 0.612 >100 6.7
154 0.592 0.984 >31.6 2.8
155 >100 5 >100 7.8
156 0.138 0.425 >100 2.1
157 >100 0.348 >100 28.5
158 >100 0.186 >100 >31.6
159 >100 0.147 >100 12.6
160 5.8 2.9 >31.6 >31.6
161 >100 1.1 >100 >100
162 >100 0.19 >100 >31.6
163 26.9 0.096 >100 >31.6
164 19.6 0.444 >100 24.3
165 19.4 1.2 >100 27.1
166 13.7 1.3 >100 21.9
167 >100 1.1 >100 27.3
168 0.15 0.612 >100 2.5
169 2.7 3.7 >100 9.5
170 0.103 2.4 >31.6 1.1
171 0.084 3 >100 1.4
172 0.41 4 >100 4
173 0.107 2.4 >100 2.9
174 4.9 3 >31.6 4.6
175 0.191 0.458 >100 2.5
177 >31.6 0.295 >100 3.9
178 >100 0.164 >31.6 2.3
179 8 0.202 1.4 8.1
180 15.7 1.1 >100 8.6
181 1.7 0.739 >100 4.3
182 >31.6 1.1 >100 21
183 0.203 2.8 >100 4.7
184 >100 0.133 >100 6.3
185 >31.6 0.154 >100 4.3
186 0.223 3.8 >100 2.5
187 16.6 0.712 >31.6 >100
188 >100 2.8 >100 >31.6
189 20.3 0.237 >100 6.2
190 >100 0.271 >100 31
192 31.6 0.422 >100 1.8
193 0.212 2.2 >100 1.5

III.2A Cellular Assay: Testing for Anti-Proliferative Effect (XTT Assay)
The principle of this test is based on the intracellular reduction of the tetrazolium dye XTT (sodium 3′-[1-(phenylaminocarbonyl)-3,4-tetrazolium]-bis(4-methoxy-6-nitro)benzene sulfonic acid, Sigma) to a formazan dye by mitochondrial dehydrogenases. The dye is only formed by metabolically active cells and its photometrically measurable intensity is a quantitative indicator for the presence of living cells. The reduction of dye formation by incubation of the cells with substances serves as a parameter for the anti-proliferative effect.
Testing
The tumour cell lines (ATCC) were injected into 96-well microtitre plates in a defined cell number (5000 cells/well for BxPC3 and Hct116; 10000 cells/well for MDA MB468) and then incubated overnight in an incubator at 37° C., 5% CO2 and 95% air humidity. The test substances were prepared as stock solutions (10 mM) in DMSO. To determine the EC50 values the potential inhibitor substances were added to the cells in quarter-logarithmically graded dilutions, resulting in final concentrations of 0.28 μM-50 μM. The cell plates were then incubated for 45 h in an incubator at 37° C., 5% CO2 and 95% air humidity.
For the detection reaction the substrate XTT was mixed with PMS (N-Methyl dibenzopyrazine methylsulfate, Sigma) and added to the cells so that a final concentration of 325 μg XTT/ml and 2.5 μg PMS/ml was obtained. It was then incubated for 3 h at 37° C., 95% air humidity. The formazan salt formed by the cellular dehydrogenases could then be quantified by adsorption at 490 nm.
Evaluation
The % inhibition value was evaluated by means of the following formula from the values for the optical densities measured in each case at 490 nm:
% Inhibition of cell proliferation ( Sample ) = 100 - ( 100 × Mean ( Sample ) - Mean ( 0 % Control ) Mean ( 100 % Control ) - Mean ( 0 % Control ) )
Eight determinations were made for each control and two for the substance samples. The 0% control contains no cells, the 100% control (proliferation control) contains no test substance. The EC50 values were determined using GraphPadPrism.
The compounds according to the invention showed partly effective inhibition of the cell proliferation with EC50 values of to <1 μM (see Table 3).
TABLE 3
XTT assay test results (EC50 [μM])
Compound BxPC3 MDA-MB468 Hct116
 30 12 4 6
124 10 10 1.5
146 >50 ca. 20 9.3
150 0.9 4.3 2.3
153 >25 24 6
156 8.9 15 1.4
157 >25 3.5 4.4
164 >50 5.2 >25
169 nicht getestet 5.7 2.8
173 nicht getestet >25 9
175 nicht getestet >50 4.4
176 nicht getestet 17 11.8
183 nicht getestet 4.3 2.1
191 nicht getestet 8.5 5.5
193 nicht getestet 4.4 2.1
Ly294002 25 20 17
It was surprisingly found that the PI3K inhibitor Ly294002 known from the literature only showed weak anti-proliferative effects on the cell lines used compared to the Exemplary Embodiments.
III.2B Cellular Assay: Testing for Substrate Inhibition (Western Blotting)
This method can be used to predict whether the kinase modulator under study also achieves the desired effect in a cellular context, i.e. in this case, a substrate protein downstream of the target kinase is investigated for its phosphorylation status. For this purpose, the cells incubated with substance are lysed and the total protein separated on a reducing polyacrylamide gel. The proteins are then transferred to a PVDF membrane by means of Western Blotting and the substrate bands sought are made visible with specific antibodies and a suitable detection method. The substrate proteins downstream of the target kinases are detected simultaneously with a respectively special anti-phospho-antibody and at the same time a total antibody which recognizes the substrate total protein. This simultaneous measurement can be made using the duplex technology of the ODYSSEY imagers (LiCOR). The intensity of the total substrate bands is used for normalising or quantifying the phosphorylation inhibition or activation.
Testing
Suitable tumour cell lines (e.g. BxPC3, Hct116 or MDA MB468) were injected into six-well microtitre plates in a defined cell number (e.g. 350 000 cells/well for BxPC3 and Hct116) in the respective standard complete medium and then incubated overnight at 37° C., 5% CO2 and 95% air humidity. The cells were then incubated for a further 24 h under serum-reduced conditions i.e. in the respective medium but with only 0.25% serum. The test substances were prepared as stock solutions (10 mM) in DMSO and incubated with the cells in final concentrations of 5, 15.8 and 50 μM for 5 h. This was followed by cell lysis in 25 mM Tris, 150 mM NaCl, 10 mM Na-pyrophosphate, 2 mM EGTA, 25 mM β-glycerophosphate, 25 mM NaF, 10% glycerine, 0.75% NP-40, 100 μM NaVO4 buffer. Following protein quantification by means of BCA (bicinchonic acid protein assay kit, Sigma) Assay, protein quantities of about 20 μg per track were separated on a Lämmli polyacrylamide gel and then transferred to a PVDF membrane (Millipore) by means of Semi-Dry Western-Blotting at 0.8 mA/cm2 for 1 h. This was followed by pre-hybridisation of the membrane for 1 hour in I-block reagent (Applied Biosystems) and incubation overnight with the specific antibodies. To determine the Erk and PI3K inhibition, the respectively downstream substrates Rsk1 were detected with the total antibody (Rsk #sc-231g C-21, Santa Cruz) and the phospho-antibody (Phospho-p90RSK (S380) #9341, NEB Cell Signalling) and Akt was detected with the total antibody (Akt1 #sc-1618 C-20, Santa Cruz) and the phospho-antibody (Phospho-Akt (Ser 473) #9271, NEB Cell Signaling). After washing the membrane, secondary antibody incubation was carried out with anti-rabbit IR Dye 800 (#611-732-127, Rockland) for the phospho-antibody and anti-goat Alexa Fluor 680 (#A-21081, Molecular Probes) for the total protein-antibody. After incubating for 30 min at room temperature in the dark, the hybridisation of the detection antibody on the membrane was detected by scanning in the ODYSSEY imager (LiCOR).
Evaluation
At concentrations of 5-50 μM, the compounds according to the invention exhibited dual inhibition of Erk (MAPK1/2) and PI3K (see Table 4) which are indicated by inhibition of the band intensity of both corresponding phospho-substrate proteins Rsk1 and Akt. The reduction of the fluorescence intensity of the phospho-substrate bands (pRsk and pAkt) is given in the table below as % inhibition and relates to the following formula:
% Inhibition of Substrate phosphorylation ( Sample ) = 100 - ( 100 × Sample 100 % Control )
The band intensity (fluorescence intensity) of the respective non-inhibited (without substance) phospho-substrates was used as 100% control.
TABLE 4
Inhibition of cellular substrate phosphorylation by selected substances
(bei 50 μM)
Compound Erk → pRsk PI3K → pAkt
 30 90% 100%
124 90% 90%
146 100% 10%
150 0% 90%
173 90% 50%
183 80% 80%
Ly294002 0% 40%
Wortmannin 0% 100%
The PI3K inhibitor Ly294002 known from the literature showed only weak PI3K inhibition compared to the pyridopyrazine derivatives, i.e. inhibition of the PI3K substrate p-Akt and as predicted, no Erk inhibition or inhibition of the Erk substrate p-Rsk. Wortmannin—a further PI3K inhibitor known from the literature—showed complete inhibition of the PI3K substrate pAkt, but no Erk or p-RSK inhibition.
In contrast to Exemplary Embodiments both reference substances used here showed no dual inhibition, i.e. of Erk and PI3K simultaneously but only a PI3K inhibition.
ABBREVIATIONS
Akt of: murine Akt8 retrovirus or protein kinase B (PKB)
Ask1 apoptosis signal-regulating kinase
ATR ataxia-telangiectasia and Rad3-related
ATM Ataxia-telangiectasia mutated
Bag1 Bcl-2 associated athanogene-1
Bcl-2 B-cell leukemia/lymphoma-2 gene
DNA-PK DNA-dependent protein kinase
Erk extracellular signal-regulated kinase
Flt-3 fms like tyrosine kinase 3
GSK-3 Glycogen synthase kinase-3
hSMG-1 human ortholog of product of seven nematode gene-1
JAK-3 Janus kinase 3
JNK c-jun N-terminal kinase
MAPK mitogen activated protein kinase
Mek MAP or Erk kinase
mTOR mammalian target of rapamycin
PDGFR platelet derived growth factor receptor
PI3K phosphoinositol 3-kinase
PIKK phosphoinositol 3-kinase related kinase
PIP2 phosphatidylinositol-biphosphate
PIP3 phosphatidylinositol-triphosphate
PtdIns phosphatidylinositol
Raf rapid accelerated fibrosarcoma
Ras rat sarcoma
RTK receptor tyrosine kinase
SAPK stress-activated protein kinase
Ser Serine
Syk spleen tyrosine kinase
Thr Threonine
Tyr Tyrosine
VEGFR vascular endothelial growth factor receptor
REFERENCES
  • 1. Alessi D R, Andjelkovic M, Caudwell B, Cron P, Morrice N, Cohen P, Hemmings B A. Mechanism of activation of protein kinase B by insulin and IGF-1. EMBO J. 1996 Dec. 2; 15(23):6541-51.
  • 2. Ali K, Bilancio A, Thomas M, Pearce W, Gilfillan A M, Tkaczyk C, Kuehn N, Gray A, Giddings J, Peskett E, Fox R, Bruce I, Walker C, Sawyer C, Okkenhaug K, Finan P, Vanhaesebroeck B. Essential role for the p110delta phosphoinositide 3-kinase in the allergic response. Nature. 2004 Oct. 21; 431(7011):1007-11.
  • 3. Bennett B L, Sasaki D T, Murray B W, O'Leary E C, Sakata S T, Xu W, Leisten J C, Motiwala A, Pierce S, Satoh Y, Bhagwat S S, Manning A M, Anderson D W. SP600125, an anthrapyrazolone inhibitor of Jun N-terminal kinase. Proc Natl Acad Sci USA. 2001 Nov. 20; 98(24):13681-6.
  • 4. Bondev A, Rubio I, Wetzker R. Differential regulation of lipid and protein kinase activities of phosphoinositide 3-kinase gamma in vitro. Biol. Chem. 1999 November; 380(11):1337-40.
  • 5. Bondeva T, Pirola L, Bulgarelli-Leva G, Rubio I, Wetzker R, Wymann M P. Bifurcation of lipid and protein kinase signals of PI3Kgamma to the protein kinases PKB and MAPK. Science. 1998 Oct. 9; 282(5387):293-6.
  • 6. Campbell I G, Russell S E, Choong D Y, Montgomery K G, Ciavarella M L, Hooi C S, Cristiano B E, Pearson R B, Phillips W A. Mutation of the PIK3CA gene in ovarian and breast cancer. Cancer Res. 2004 Nov. 1; 64(21):7678-81.
  • 7. Chang F, Lee J T, Navolanic P M, Steelman L S, Shelton J G, Blalock W L, Franklin R A, McCubrey J A. Involvement of PI3K/Akt pathway in cell cycle progression, apoptosis, and neoplastic transformation: a target for cancer chemotherapy. Leukemia. 2003 March; 17(3):590-603. Review
  • 8. Chang F, Steelman L S, Lee J T, Shelton J G, Navolanic P M, Blalock W L, Franklin R A, McCubrey J A. Signal transduction mediated by the Ras/Raf/MEK/ERK pathway from cytokine receptors to transcription factors: potential targeting for therapeutic intervention. Leukemia. 2003 July; 17(7):1263-93. Review.
  • 9. Chang H W, Aoki M, Fruman D, Auger K R, Bellacosa A, Tsichlis P N, Cantley L C, Roberts T M, Vogt P K. Transformation of chicken cells by the gene encoding the catalytic subunit of PI 3-kinase. Science. 1997 Jun. 20; 276(5320):1848-50.
  • 10. Chen J, Fujii K, Zhang L, Roberts T, Fu H. Raf-1 promotes cell survival by antagonizing apoptosis signal-regulating kinase 1 through a MEK-ERK independent mechanism. Proc Natl Acad Sci USA. 2001 Jul. 3; 98(14):7783-8.
  • 11. Chiang G G, Abraham R T. Determination of the catalytic activities of mTOR and other members of the phosphoinositide-3-kinase-related kinase family. Methods Mol Biol. 2004; 281:125-41.
  • 12. Crackower M A, Oudit G Y, Kozieradzki I, Sarao R, Sun H, Sasaki T, Hirsch E, Suzuki A, Shioi T, Irie-Sasaki J, Sah R, Cheng H Y, Rybin V O, Lembo G, Fratta L, Oliveira-dos-Santos A J, Benovic J L, Kahn C R, Izumo S, Steinberg S F, Wymann M P, Backx P H, Penninger J M. Regulation of myocardial contractility and cell size by distinct PI3K-PTEN signaling pathways. Cell. 2002 Sep. 20; 110(6):737-49.
  • 13. Davies H, Bignell G R, Cox C, Stephens P, Edkins S, Clegg S, Teague J, Woffendin H, Garnett M J, Bottomley W, Davis N, Dicks E, Ewing R, Floyd Y, Gray K, Hall S, Hawes R, Hughes J, Kosmidou V, Menzies A, Mould C, Parker A, Stevens C, Watt S, Hooper S, Wilson R, Jayatilake H, Gusterson B A, Cooper C, Shipley J, Hargrave D, Pritchard-Jones K, Maitland N, Chenevix-Trench G, Riggins G J, Bigner D D, Palmieri G, Cossu A, Flanagan A, Nicholson A, Ho J W, Leung S Y, Yuen S T, Weber B L, Seigler H F, Darrow T L, Paterson H, Marais R, Marshall C J, Wooster R, Stratton M R, Futreal P A. Mutations of the BRAF gene in human cancer. Nature. 2002 Jun. 27; 417(6892):949-54.
  • 14. Davies S P, Reddy H, Caivano M, Cohen P. Specificity and mechanism of action of some commonly used protein kinase inhibitors. Biochem J. 2000 Oct. 1; 351 (Pt 1):95-105.
  • 15. Dhand R, Hiles I, Panayotou G, Roche S, Fry M J, Gout I, Totty N F, Truong O, Vicendo P, Yonezawa K, et al. PI 3-kinase is a dual specificity enzyme: autoregulation by an intrinsic protein-serine kinase activity. EMBO J. 1994 Feb. 1; 13(3):522-33.
  • 16. Elliott R D, Temple C Jr, Montgomery J A. Potential folic acid antagonists. 3. Deaza analogs of methotrexate. 3. 1- and 3-Deaza analogs of 2,4-diamino-6-[(n-methylanilino)methyl]pteridine. J Org Chem. 1968 February; 33(2):533-6.
  • 17. Friess H, Berberat P, Schilling M, Kunz J, Korc M, Buchler M W. Pancreatic cancer: the potential clinical relevance of alterations in growth factors and their receptors. J Mol Med. 1996 January; 74(1):35-42. Review.
  • 18. Gotz R, Wiese S, Takayama S, Camarero G C, Rossoll W, Schweizer U, Troppmair J, Jablonka S, Holtmann B, Reed J C, Rapp U R, Sendtner M. Bag1 is essential for differentiation and survival of hematopoietic and neuronal cells. Nat. Neurosci. 2005 September; 8(9): 1169-78.
  • 19. Gum R J, McLaughlin M M, Kumar S, Wang Z, Bower M J, Lee J C, Adams J L, Livi G P, Goldsmith E J, Young P R. Acquisition of sensitivity of stress-activated protein kinases to the p38 inhibitor, SB 203580, by alteration of one or more amino acids within the ATP binding pocket. J Biol Chem. 1998 Jun. 19; 273(25):15605-10.
  • 20. Hoshino R, Chatani Y, Yamori T, Tsuruo T, Oka H, Yoshida O, Shimada Y, Ari-i S, Wada H, Fujimoto J, Kohno M. Constitutive activation of the 41-/43-kDa mitogen-activated protein kinase signaling pathway in human tumors. Oncogene. 1999 Jan. 21; 18(3):813-22.
  • 21. Katso R, Okkenhaug K, Ahmadi K, White S, Timms J, Waterfield M D. Cellular function of phosphoinositide 3-kinases: implications for development, homeostasis, and cancer. Annu Rev Cell Dev Biol. 2001; 17:615-75. Review.
  • 22. Khleif S N, Abrams S I, Hamilton J M, Bergmann-Leitner E, Chen A, Bastian A, Bernstein S, Chung Y, Allegra C J, Schlom J. A phase I vaccine trial with peptides reflecting ras oncogene mutations of solid tumors. J Immunother. 1999 March; 22(2): 155-65.
  • 23. Levine D A, Bogomolniy F, Yee C J, Lash A, Barakat R R, Borgen P I, Boyd J. Frequent mutation of the PIK3CA gene in ovarian and breast cancers. Clin Cancer Res. 2005 Apr. 15; 11(8):2875-8.
  • 24. Lewis T S, Shapiro P S, Ahn N G. Signal transduction through MAP kinase cascades. Adv Cancer Res. 1998; 74:49-139. Review.
  • 25. Li J, Yen C, Liaw D, Podsypanina K, Bose S, Wang S I, Puc J, Miliaresis C, Rodgers L, McCombie R, Bigner S H, Giovanella B C, Ittmann M, Tycko B, Hibshoosh H, Wigler M H, Parsons R. PTEN, a putative protein tyrosine phosphatase gene mutated in human brain, breast, and prostate cancer. Science. 1997 Mar. 28; 275(5308):1943-7.
  • 26. Lu Y, Wang H, Mills G B. Targeting PI3K-AKT pathway for cancer therapy. Rev Clin Exp Hematol. 2003 June; 7(2):205-28. Review.
  • 27. Ma Y Y, Wei S J, Lin Y C, Lung J C, Chang T C, Whang-Peng J, Liu J M, Yang D M, Yang W K, Shen C Y. PIK3CA as an oncogene in cervical cancer. Oncogene. 2000 May 25; 19(23):2739-44.
  • 28. Marshall C. How do small GTPase signal transduction pathways regulate cell cycle entry? Curr Opin Cell Biol. 1999 December; 11(6):732-6. Review.
  • 29. McPhillips F, Mullen P, Monia B P, Ritchie A A, Dorr F A, Smyth J F, Langdon S P. Association of c-Raf expression with survival and its targeting with antisense oligonucleotides in ovarian cancer. Br J Cancer. 2001 Nov. 30; 85(11):1753-8.
  • 30. Mendelsohn J, Baselga J. The EGF receptor family as targets for cancer therapy. Oncogene. 2000 Dec. 27; 19(56):6550-65. Review.
  • 31. Moore S M, Rintoul R C, Walker T R, Chilvers E R, Haslett C, Sethi T. The presence of a constitutively active phosphoinositide 3-kinase in small cell lung cancer cells mediates anchorage-independent proliferation via a protein kinase B and p70s6k-dependent pathway. Cancer Res. 1998 Nov. 15; 58(22):5239-47.
  • 32. Okkenhaug K, Vanhaesebroeck B. PI3K in lymphocyte development, differentiation and activation. Nat Rev Immunol. 2003 April; 3(4):317-30. Review.
  • 33. Patrucco E, Notte A, Barberis L, Selvetella G, Maffei A, Brancaccio M, Marengo S, Russo G, Azzolino O, Rybalkin S D, Silengo L, Altruda F, Wetzker R, Wymann M P, Lembo G, Hirsch E. PI3Kgamma modulates the cardiac response to chronic pressure overload by distinct kinase-dependent and -independent effects. Cell. 2004 Aug. 6; 118(3):375-87.
  • 34. Rapp U R, Rennefahrt U, Troppmair J. Bcl-2 proteins: master switches at the intersection of death signaling and the survival control by Raf kinases. Biochim Biophys Acta. 2004 Mar. 1; 1644(2-3):149-58. Review.
  • 35. Rodriguez-Viciana P, Warne P H, Dhand R, Vanhaesebroeck B, Gout I, Fry M J, Waterfield M D, Downward J. Phosphatidylinositol-3-OH kinase as a direct target of Ras. Nature. 1994 Aug. 18; 370(6490):527-32.
  • 36. Samuels Y, Wang Z, Bardelli A, Silliman N, Ptak J, Szabo S, Yan H, Gazdar A, Powell S M, Riggins G J, Willson J K, Markowitz S, Kinzler K W, Vogelstein B, Velculescu V E. High frequency of mutations of the PIK3CA gene in human cancers, Science. 2004 Apr. 23; 304(5670):554.
  • 37. Sebolt-Leopold J S, Herrera R. Targeting the mitogen-activated protein kinase cascade to treat cancer. Nat Rev Cancer. 2004 December; 4(12):937-47. Review.
  • 38. Shayesteh L, Lu Y, Kuo W L, Baldocchi R, Godfrey T, Collins C, Pinkel D, Powell B, Mills G B, Gray J W. PIK3CA is implicated as an oncogene in ovarian cancer Nat Genet. 1999 January; 21(1):99-102.
  • 39. Sirivatanauksorn V, Sirivatanauksorn Y, Lemoine N R. Molecular pattern of ductal pancreatic cancer. Langenbecks Arch Surg. 1998 April; 383(2):105-15. Review.
  • 40. Steck P A, Pershouse M A, Jasser S A, Yung W K, Lin H, Ligon A H, Langford L A, Baumgard M L, Hattier T, Davis T, Frye C, Hu R, Swedlund B, Teng D H, Tavtigian S V. Identification of a candidate tumour suppressor gene, MMAC 1, at chromosome 10q23.3 that is mutated in multiple advanced cancers. Nat Genet. 1997 April; 15(4):356-62.
  • 41. Sujobert P, Bardet V, Cornillet-Lefebvre P, Hayflick J S, Prie N, Verdier F, Vanhaesebroeck B, Muller O, Pesce F, Ifrah N, Hunault-Berger M, Berthou C, Villemagne B, Jourdan E, Audhuy B, Solary E, Witz B, Harousseau J L, Himberlin C, Lamy T, Lioure B, Cahn J Y, Dreyfus F, Mayeux P, Lacombe C, Bouscary D. Essential role for the p110delta isoform in phosphoinositide 3-kinase activation and cell proliferation in acute myeloid leukemia. Blood. 2005 Aug. 1; 106(3):1063-6.
  • 42. Temple C Jr, Rener G A. Potential antimitotic agents. Synthesis of some ethyl benzopyrazin-7-ylcarbamates, ethyl pyrido[3,4-b]pyrazin-7-ylcarbamates, and ethyl pyrido[3,4-e]-as-triazin-7-ylcarbamates. J Med Chem. 1990 November; 33(11):3044-50.
  • 43. Troppmair J, Rapp U R. Raf and the road to cell survival: a tale of bad spells, ring bearers and detours. Biochem Pharmacol. 2003 Oct. 15; 66(8):1341-5. Review.
  • 44. Vanhaesebroeck B, Higashi K, Raven C, Welham M, Anderson S, Brennan P, Ward S G, Waterfield M D. Autophosphorylation of p110delta phosphoinositide 3-kinase: a new paradigm for the regulation of lipid kinases in vitro and in vivo. EMBO J. 1999 Mar. 1; 18(5):1292-302.
  • 45. Vanhaesebroeck B, Leevers S J, Ahmadi K, Timms J, Katso R, Driscoll P C, Woscholski R, Parker P J, Waterfield M D. Synthesis and function of 3-phosphorylated inositol lipids. Annu Rev Biochem. 2001; 70:535-602. Review.
  • 46. Weinstein-Oppenheimer C R, Blalock W L, Steelman L S, Chang F, McCubrey J A. The Raf signal transduction cascade as a target for chemotherapeutic intervention in growth factor-responsive tumors. Pharmacol Ther. 2000 December; 88(3):229-79. Review.
  • 47. Wetzker R, Rommel C. Phosphoinositide 3-kinases as targets for therapeutic intervention. Curr Pharm Des. 2004; 10(16):1915-22. Review.
  • 48. Wymann M P, Pirola L. Structure and function of phosphoinositide 3-kinases. Biochim Biophys Acta. 1998 Dec. 8; 1436(1-2):127-50. Review.

Claims (22)

The invention claimed is:
1. A method for treating a mammal having a malignant tumor comprising:
administering to a mammal in need thereof an effective amount of at least one compound according to general formula (I):
Figure US08937068-20150120-C00054
wherein the substituents R1, R2, R3, R4 have the following meanings:
R1 is NR7R8,
wherein R7 and R8 independently of each other are hydrogen, alkyl, cycloalkyl, aryl, heteroaryl, alkyl-cycloalkyl, alkyl-heterocyclyl, alkyl-aryl or alkyl-heteroaryl; wherein said alkyl-, cycloalkyl-, aryl- and heteroaryl-, alkyl-cycloalkyl, alkyl-heterocyclyl, alkyl-aryl or alkyl-heteroaryl substituents are independently of each other optionally substituted by F, Cl, Br, I, CN, CF3, NH2, NH-alkyl, NH-aryl, N(alkyl)2, NO2, SH, S-alkyl, OH, OCF3, O-alkyl, O-aryl, OSO2H, OP(O)(OH)2, CHO, CO2H, SO3H or alkyl, and wherein R7 and R8 do not together form a heterocycle,
R2 is hydrogen,
R3 is —NR9R10,
wherein R9 is hydrogen or alkyl and R10 is —C(Y)NR11R12, wherein Y═O or S and R11 and R12 are independently of each other
(i) hydrogen,
(ii) unsubstituted or substituted alkyl, wherein the alkyl group is optionally substituted with one or more substitutents selected from the group consisting of F, Cl, Br, I, CF3, CN, NH2, NH-alkyl, NH-cycloalkyl, NH-heterocyclyl, NH-aryl, NH-heteroaryl, NH-alkyl-cycloalkyl, NH-alkyl-heterocyclyl, NH-alkyl-aryl, NH-alkyl-heteroaryl, N(alkyl)2, NHC(O)-alkyl, NHC(O)-cycloalkyl, NHC(O)-heterocyclyl, NHC(O)-aryl, NHC(O)-heteroaryl, NHC(O)-alkyl-aryl, NHC(O)-alkyl-heteroaryl, NHSO2-alkyl, NHSO2-cycloalkyl, NHSO2-heterocyclyl, NHSO2-aryl, NHSO2-heteroaryl, NHSO2-alkyl-aryl, NHSO2-alkyl-heteroaryl, NO2, SH, S-alkyl, S-cycloalkyl, S-heterocyclyl, S-aryl, S-heteroaryl, OH, OCF3, O-alkyl, O-cycloalkyl, O-heterocyclyl, O-aryl, O-heteroaryl, O-alkyl-cycloalkyl, O-alkyl-heterocyclyl, O-alkyl-aryl, O-alkyl-heteroaryl, OC(O)-alkyl, OC(O)cycloalkyl, OC(O)-heterocyclyl, OC(O)-aryl, OC(O)-heteroaryl, OC(O)-alkyl-aryl, OC(O)alkyl-heteroaryl, OSO3H, OSO2-alkyl, OSO2-cycloalkyl, OSO2-heterocyclyl, OSO2-aryl, OSO2-heteroaryl, OSO2-alkyl-aryl, OSO2-alkyl-heteroaryl, OP(O)(OH)2, C(O)-alkyl, C(O)-aryl, C(O)-heteroaryl, CO2H, CO2-alkyl, CO2-cycloalkyl, CO2-heterocyclyl, CO2-aryl, CO2-heteroaryl, CO2-alkyl-cycloalkyl, CO2-alkyl-heterocyclyl, CO2-alkyl-aryl, CO2-alkyl-heteroaryl, C(O)—NH2, C(O)NH-alkyl, C(O)NH-cycloalkyl, C(O)NH-heterocyclyl, C(O)NH-aryl, C(O)NH-heteroaryl, C(O)NH-alkyl-cycloalkyl, C(O)NH-alkyl-heterocyclyl, C(O)NH-alkyl-aryl, C(O)NH-alkyl-heteroaryl, C(O)N(alkyl)2, C(O)N(cycloalkyl)2, C(O)N(aryl)2, C(O)N(heteroaryl)2, SO-alkyl, SO-aryl, SO2-alkyl, SO2-aryl, SO2NH2, SO2NH-alkyl, SO2NH-aryl, SO2NH-heteroaryl, SO2NH-alkyl-aryl, SO3H, SO2O-alkyl, SO2O-aryl, SO2O-alkyl-aryl, cycloalkyl, heterocyclyl, aryl and heteroaryl, which may be the same or different from each other;
(iii) unsubstituted or substituted cycloalkyl, wherein the cycloalkyl group is optionally substituted with one or more substituents selected from the group consisting of F, Cl, Br, I, NH2, NH-alkyl, NH-cycloalkyl, NH-heterocyclyl, NH-aryl, NH-heteroaryl, NH-alkyl-aryl, NH-alkyl-heteroaryl, N(alkyl)2, NHC(O)-alkyl, NHC(O)cycloalkyl, NHC(O)-heterocyclyl, NHC(O)-aryl, NHC(O)-heteroaryl, NHC(O)-alkyl-aryl, NHC(O)-alkyl-heteroaryl, NHSO2-alkyl, NHSO2-cycloalkyl, NHSO2-heterocyclyl, NHSO2-aryl, NHSO2-heteroaryl, NHSO2-alkyl-aryl, NHSO2-alkyl-heteroaryl, OH, O-alkyl, O-cycloalkyl, O-heterocyclyl, O-aryl, O-heteroaryl, O-alkyl-aryl, O-alkyl-heteroaryl, OC(O)alkyl, OC(O)-cycloalkyl, OC(O)-heterocyclyl, OC(O)-aryl, OC(O)-heteroaryl, OC(O)-alkyl-aryl, OC(O)-alkyl-heteroaryl, OSO3H, OSO2-alkyl, OSO2-cycloalkyl, OSO2-heterocyclyl, OSO2-aryl, OSO2-heteroaryl, OSO2-alkyl-aryl, OSO2-alkyl-heteroaryl, OP(O)(OH)2, CO2H, CO2-alkyl, CO2-cycloalkyl, CO2-heterocyclyl, CO2-aryl, CO2-heteroaryl, CO2-alkyl-cycloalkyl, CO2-alkyl-heterocyclyl, CO2-alkyl-aryl, CO2-alkyl-heteroaryl, C(O)—NH2, C(O)NH-alkyl, C(O)NH-cycloalkyl, C(O)NH-heterocyclyl, C(O)NH-aryl, C(O)NH-heteroaryl, C(O)NH-alkyl-cycloalkyl, C(O)NH-alkyl-heterocyclyl, C(O)NH-alkyl-aryl, C(O)NH-alkyl-heteroaryl, C(O)N(alkyl)2, C(O)N(cycloalkyl)2, C(O)N(aryl)2, C(O)N(heteroaryl)2, alkyl, and aryl, which may be the same or different from each other;
(iv) unsubstituted or substituted heterocyclyl, wherein the heterocyclyl group is optionally substituted with one or more substituents selected from the group consisting of OH, O-alkyl, O-aryl, NH2, NH-alkyl, NH-aryl, alkyl, alkyl-aryl or and aryl, which may be the same or different from each other;
(v) unsubstituted or substituted aryl, wherein the aryl group is optionally substituted with one or more substitutents selected from the group consisting of F, Cl, Br, I, CF3, CN, NH2, NH-alkyl, NH-cycloalkyl, NH-heterocyclyl, NH-aryl, NH-heteroaryl, NH-alkyl-cycloalkyl, NH-alkyl-heterocyclyl, NH-alkyl-aryl, NH-alkyl-heteroaryl, NH-alkyl-NH2, NH-alkyl-OH, N(alkyl)2, NHC(O)-alkyl, NHC(O)-cycloalkyl, NHC(O)-heterocyclyl, NHC(O)-aryl, NHC(O)-heteroaryl, NHC(O)-alkyl-aryl, NHC(O)-alkyl-heteroaryl, NHSO2-alkyl, NHSO2-cycloalkyl, NHSO2-heterocyclyl, NHSO2-aryl, NHSO2-heteroaryl, NHSO2-alkyl-aryl, NHSO2-alkyl-heteroaryl, NO2, SH, S-alkyl, S-cycloalkyl, S-heterocyclyl, S-aryl, S-heteroaryl, OH, OCF3, O-alkyl, O-cycloalkyl, O-heterocyclyl, O-aryl, O-heteroaryl, O-alkyl-cycloalkyl, O-alkyl-heterocyclyl, O-alkyl-aryl, O-alkyl-heteroaryl, O-alkyl-OH, O(CH2)n—O, OC(O)-alkyl, OC(O)-cycloalkyl, OC(O)-heterocyclyl, OC(O)-aryl, OC(O)heteroaryl, OC(O)-alkyl-aryl, OC(O)-alkyl-heteroaryl, OSO3H, OSO2-alkyl, OSO2-cycloalkyl, OSO2-heterocyclyl, OSO2-aryl, OSO2-heteroaryl, OSO2-alkyl-aryl, OSO2-alkyl-heteroaryl, OP(O)(OH)2, C(O)-alkyl, C(O)-aryl, C(O)-heteroaryl, CO2H, CO2-alkyl, CO2-cycloalkyl, CO2-heterocyclyl, CO2-aryl, CO2-heteroaryl, CO2-alkyl-cycloalkyl, CO2-alkyl-heterocyclyl, CO2-alkyl-aryl, CO2-alkyl-heteroaryl, C(O)—NH2, C(O)NH-alkyl, C(O)NH-cycloalkyl, C(O)NH-heterocyclyl, C(O)NH-aryl, C(O)NH-heteroaryl, C(O)NH-alkyl-cycloalkyl, C(O)NH-alkyl-heterocyclyl, C(O)NH-alkyl-aryl, C(O)NH-alkyl-heteroaryl, C(O)N(alkyl)2, C(O)N(cycloalkyl)2, C(O)N(aryl)2, C(O)N(heteroaryl)2, SO-alkyl, SO-aryl, SO2-alkyl, SO2-aryl, SO2NH2, SO2NH-alkyl, SO2NH-aryl, SO2NH-heteroaryl, SO2NH-alkyl-aryl, SO3H, SO2O-alkyl, SO2O-aryl, SO2O-alkyl-aryl, alkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl, which may be the same or different from each other, and n can have the value 1, 2 or 3;
(vi) unsubstituted or substituted heteroaryl, wherein the heteroaryl group is optionally substituted with one or more substituents selected from the group consisting of F, Cl, Br, I, CF3, CN, NH2, NH-alkyl, NH-cycloalkyl, NH-heterocyclyl, NH-aryl, NH-heteroaryl, NH-alkyl-cycloalkyl, NH-alkyl-heterocyclyl, NH-alkyl-aryl, NH-alkyl-heteroaryl, NH-alkyl-NH2, NH-alkyl-OH, N(alkyl)2, NHC(O)-alkyl, NHC(O)-cycloalkyl, NHC(O)-heterocyclyl, NHC(O)-aryl, NHC(O)-heteroaryl, NHC(O)-alkyl-aryl, NHC(O)-alkyl-heteroaryl, NHSO2-alkyl, NHSO2-cycloalkyl, NHSO2-heterocyclyl, NHSO2-aryl, NHSO2-heteroaryl, NHSO2-alkyl-aryl, NHSO2-alkyl-heteroaryl, NO2, SH, S-alkyl, S-aryl, S-heteroaryl, OH, OCF3, O-alkyl, O-cycloalkyl, O-aryl, O-heteroaryl, O-alkyl-cycloalkyl, O-alkyl-heterocyclyl, O-alkyl-aryl, O-alkyl-heteroaryl, OC(O)-alkyl, OC(O)-cycloalkyl, OC(O)-heterocyclyl, OC(O)-aryl, OC(O)-heteroaryl, OC(O)-alkyl-aryl, OC(O)-alkyl-heteroaryl, OSO3H, OSO2-alkyl, OSO2-cycloalkyl, OSO2-heterocyclyl, OSO2-aryl, OSO2-heteroaryl, OSO2-alkyl-aryl, OSO2-alkyl-heteroaryl, OP(O)(OH)2, C(O)-alkyl, C(O)-aryl, C(O)-heteroaryl, CO2H, CO2-alkyl, CO2-cycloalkyl, CO2-heterocyclyl, CO2-aryl, CO2-heteroaryl, CO2-alkyl-cycloalkyl, CO2-alkyl-heterocyclyl, CO2-alkyl-aryl, CO2-alkyl-heteroaryl, C(O)—NH2, C(O)NH-alkyl, C(O)NH-cycloalkyl, C(O)NH-heterocyclyl, C(O)NH-aryl, C(O)NH-heteroaryl, C(O)NH-alkyl-cycloalkyl, C(O)NH-alkyl-heterocyclyl, C(O)NH-alkyl-aryl, C(O)NH-alkyl-heteroaryl, C(O)N(alkyl)2, C(O)N(cycloalkyl)2, C(O)N(aryl)2, C(O)N(heteroaryl)2, SO2NH2, SO2NH-alkyl, SO2NH-aryl, SO2NH-heteroaryl, SO2NH-alkyl-aryl, SO3H, SO2O-alkyl, SO2O-aryl, SO2O-alkyl-aryl, alkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl, which may be the same or different from each other;
(vii) —C(O)—R17, wherein R17 is alkyl, aryl or heteroaryl, wherein the alkyl and aryl substituents are optionally substituted by F, Cl, Br, I, CN, CF3, NH2, NH-alkyl, NH-aryl, N(alkyl)2, NO2, SH, S-alkyl, OH, OCF3, O-alkyl, O-aryl, OSO2H, OP(O)(OH)2, CHO, CO2H, SO3H or alkyl; and
(viii) or R11 and R12 together form heterocyclyl,
and R4 is hydrogen;
wherein said compound inhibits PI3K alpha, PI3K beta, PI3K gamma, and PI3K delta in the PI3K-Akt signal transduction pathway; and
wherein said malignant tumor is selected from the group consisting of a colon tumor, a gastric tumor, an intestinal tumor, a pulmonary tumor, a pancreatic tumor, an ovarian tumor, a prostatic tumor, melanoma, a hepatic tumor, a renal tumor, a head tumor, glioma, a breast tumor, cervico-uterine carcinoma, adeno-acanthoma, an endometrial cancer, a colorectal tumor, esophageal cancer, thyroid cancer, lymphoma, and leukemia.
2. The method according to claim 1, wherein R7, R8, R9, R11, or R12 is an alkyl group that is selected from the group consisting of methyl, ethyl, n-propyl, 2-propyl, n-butyl, sec-butyl, tert-butyl, n-pentyl, iso-pentyl, neo-pentyl, n-hexyl, 2-hexyl, n-octyl, ethylenyl (vinyl), ethynyl, propenyl, propynyl, butenyl, butynyl, pentenyl, pentynyl, hexenyl, hexynyl, heptenyl, heptynyl, octenyl and octynyl.
3. The method according to claim 1, wherein R7, R8, R11, or R12 is a heteroaryl group that is selected from the group consisting of pyrrolyl, furyl, thienyl, thiazolyl, oxazolyl, isoxazolyl, pyrazolyl, imidazolyl, pyridinyl, pyrimidinyl, pyridazinyl, pyrazinyl, phthalazinyl, indolyl, indazolyl, indolizinyl, quinolinyl, isoquinolinyl, quinoxalinyl, quinazolinyl, carbazolyl, phenazinyl, phenothiazinyl and acridinyl.
4. The method according to claim 1, wherein R11 or R12 is a heterocyclyl group or R11 and R12 together form heterocyclyl group that is selected from the group consisting of tetrahydrofuryl, tetrahydropyranyl, pyrrolidinyl, piperidinyl, piperazinyl and morpholinyl.
5. The method according to claim 1, wherein said compound is selected from the group consisting of
62
Figure US08937068-20150120-C00055
1-Ethyl-3-[3-(2-methoxy-ethylamino)- pyrido[2,3-b]pyrazin-6-yl]-urea,
65
Figure US08937068-20150120-C00056
1-[3-(Cyclopropylmethyl-amino)- pyrido[2,3-b]pyrazin-6-yl]-3-ethyl-urea,
66
Figure US08937068-20150120-C00057
1-Ethyl-3-{3-[(pyridin-4-ylmethyl)- amino]-pyrido[2,3-b]pyrazin-6-yl}-urea,
83
Figure US08937068-20150120-C00058
1-(3-Amino-pyrido[2,3-b]pyrazin-6-yl)-3- ethyl-urea,
150
Figure US08937068-20150120-C00059
1-Ethyl-3-(3-p-tolylamino-pyrido[2,3- b]pyrazin-6-yl)-urea,
157
Figure US08937068-20150120-C00060
1-Ethyl-3-(3-m-tolylamino-pyrido-[2,3- b]pyrazin-6-yl)-urea,
158
Figure US08937068-20150120-C00061
1-Ethyl-3-[3-(4-methoxy-phenylamino)- pyrido[2,3-b]pyrazin-6-yl]-urea,
159
Figure US08937068-20150120-C00062
1-[3-(4-Chloro-phenylamino)-pyrido- [2,3-b]pyrazin-6-yl]-3-ethyl-urea,
160
Figure US08937068-20150120-C00063
1-Ethyl-3-(3-o-tolylamino-pyrido[2,3-b]- pyrazin-6-yl)-urea,
161
Figure US08937068-20150120-C00064
1-Ethyl-3-[3-(pyridin-3-ylamino)-pyrido- [2,3-b]pyrazin-6-yl]-urea,
162
Figure US08937068-20150120-C00065
1-Ethyl-3-[3-(4-ethyl-phenylamino)- pyrido[2,3-b]pyrazin-6-yl]-urea,
163
Figure US08937068-20150120-C00066
1-Ethyl-3-[3-(3-methoxy-4-methyl- phenylamino)-pyrido[2,3-b]pyrazin-6-yl]- urea,
164
Figure US08937068-20150120-C00067
1-Ethyl-3-[3-(4-hydroxy-phenylamino)- pyrido[2,3-b]pyrazin-6-yl]-urea,
165
Figure US08937068-20150120-C00068
1-Ethyl-3-[3-(5-methyl-pyridin-2-yl- amino)-pyrido[2,3-b]pyrazin-6-yl]-urea,
166
Figure US08937068-20150120-C00069
1-Ethyl-3-[3-(1-methyl-1H-pyrazol-3- ylamino)-pyrido[2,3-b]pyrazin-6-yl]-urea,
167
Figure US08937068-20150120-C00070
1-Ethyl-3-[3-(4-fluoro-phenylamino)- pyrido[2,3-b]pyrazin-6-yl]-urea,
177
Figure US08937068-20150120-C00071
1-[3-(3-Chloro-4-methoxy-phenylamino)- pyrido[2,3-b]pyrazin-6-yl]-3-ethyl-urea,
178
Figure US08937068-20150120-C00072
1-Ethyl-3-[3-(naphthalin-2-ylamino)- pyrido[2,3-b]pyrazin-6-yl]-urea,
179
Figure US08937068-20150120-C00073
1-Ethyl-3-[3-(quinolin-3-ylamino)- pyrido[2,3-b]pyrazin-6-yl]-urea,
180
Figure US08937068-20150120-C00074
1-[3-(3,5-Dimethoxy-phenylamino)- pyrido[2,3-b]pyrazin-6-yl]-3-ethyl-urea,
181
Figure US08937068-20150120-C00075
1-Ethyl-3-[3-(pyrazin-2-ylamino)- pyrido[2,3-b]pyrazin-6-yl]-urea,
182
Figure US08937068-20150120-C00076
1-Ethyl-3-[3-(3-isopropoxy- phenylamino)-pyrido[2,3-b]pyrazin-6-yl]- urea,
184
Figure US08937068-20150120-C00077
1-[3-(2-Chloro-pyridin-4-ylamino)- pyrido[2,3-b]pyrazin-6-yl]-3-ethyl-urea,
185
Figure US08937068-20150120-C00078
1-[3-(3,5-Dichloro-4-hydroxy- phenylamino)-pyrido[2,3-b]pyrazin-6-yl]- 3-ethyl-urea,
187
Figure US08937068-20150120-C00079
1-[3-(3,4-Dimethoxy-phenylamino)- pyrido[2,3-b]pyrazin-6-yl]-3-ethyl-urea,
188
Figure US08937068-20150120-C00080
1-Ethyl-3-[3-(3-hydroxy-4-methyl- phenylamino)-pyrido[2,3-b]pyrazin-6-yl]- urea,
189
Figure US08937068-20150120-C00081
3-[6-(3-Ethyl-ureido)-pyrido[2,3-b]- pyrazin-3-ylamino]-5-trifluoromethyl- benzoic acid,
190
Figure US08937068-20150120-C00082
1-Ethyl-3-[3-(6-methoxy-pyridin-3- ylamino)-pyrido[2,3-b]pyrazin-6-yl]-urea,
191
Figure US08937068-20150120-C00083
1-[3-(3,5-Dimethyl-phenylamino)- pyrido[2,3-b]pyrazin-6-yl]-3-ethyl-urea,
192
Figure US08937068-20150120-C00084
1-[3-(4-Cyano-phenylamino)-pyrido-[2,3- b]pyrazin-6-yl]-3-ethyl-urea,
197
Figure US08937068-20150120-C00085
1-Ethyl-3-[3-(4-hydroxy-3-methoxy- phenylamino)-pyrido[2,3-b]pyrazin-6-yl]- urea,
211
Figure US08937068-20150120-C00086
1-Ethyl-3-[3-(methyl-p-tolyl-amino)- pyrido[2,3-b]pyrazin-6-yl]-urea,
212
Figure US08937068-20150120-C00087
1-Ethyl-3-[3-(2-p-tolyl-ethylamino)- pyrido[2,3-b]pyrazin-6-yl]-urea,
213
Figure US08937068-20150120-C00088
1-Ethyl-3-[3-(4-methyl-benzylamino)- pyrido[2,3-b]pyrazin-6-yl]-urea,
214
Figure US08937068-20150120-C00089
1-Ethyl-3-[3-(3-fluoro-4-methyl-phenyl- amino)-pyrido[2,3-b]pyrazin-6-yl]-urea,
215
Figure US08937068-20150120-C00090
1-[3-(3,4-Dimethyl-phenylamino)- pyrido[2,3-b]pyrazin-6-yl]-3-ethyl-urea,
216
Figure US08937068-20150120-C00091
1-Ethyl-3-[3-(4-isopropyl-phenylamino)- pyrido[2,3-b]pyrazin-6-yl]-urea,
240
Figure US08937068-20150120-C00092
1-[3-(Benzothiazol-6-ylamino)- pyrido[2,3-b]pyrazin-6-yl]-3-ethyl-urea,
241
Figure US08937068-20150120-C00093
1-Ethyl-3-[3-(4-methyl-3- trifluoroomethyl-phenylamino)- pyrido[2,3-b]pyrazin-6-yl]-urea,
242
Figure US08937068-20150120-C00094
1-[3-(3-Cyano-4-methyl-phenylamino)- pyrido[2,3-b]pyrazin-6-yl]-3-ethyl-urea,
243
Figure US08937068-20150120-C00095
1-Ethyl-3-[3-(4-phenoxy-phenylamino)- pyrido[2,3-b]pyrazin-6-yl]-urea,
244
Figure US08937068-20150120-C00096
1-[3-(4-Chloro-3-methyl-phenylamino)- pyrido[2,3-b]pyrazin-6-yl]-3-ethyl-urea,
245
Figure US08937068-20150120-C00097
1-[3-(2-Chloro-4-methyl-phenylamino)- pyrido[2,3-b]pyrazin-6-yl]-3-ethyl-urea,
246
Figure US08937068-20150120-C00098
1-Ethyl-3-[3-(3-trifluoromethyl- phenylamino)-pyrido[2,3-b]pyrazin-6-yl]- urea,
247
Figure US08937068-20150120-C00099
1-[3-(2-Chloro-4-trifluoromethoxy- phenylamino)-pyrido[2,3-b]pyrazin-6-yl]- 3-ethyl-urea,
248
Figure US08937068-20150120-C00100
1-[3-(4-Chloro-2-methoxy-5-methyl- phenylamino)-pyrido[2,3-b]pyrazin-6-yl]- 3-ethyl-urea,
252
Figure US08937068-20150120-C00101
1-[3-(Benzo[1,3]-dioxol-5-ylamino)- pyrido[2,3-b]pyrazin-6-yl]-3-ethyl-urea,
253
Figure US08937068-20150120-C00102
3-[6-(3-Ethyl-ureido)-pyrido[2,3- b]pyrazin-3-ylamino]-benzoic acid,
254
Figure US08937068-20150120-C00103
2-Chloro-4-[6-(3-ethyl-ureido)- pyrido[2,3-b]pyrazin-3-ylamino]-benzoic acid,
255
Figure US08937068-20150120-C00104
1-Ethyl-3-[3-(3-methoxy-5- trifluoromethyl-phenylamino)-pyrido[2,3- b]pyrazin-6-yl]-urea,
256
Figure US08937068-20150120-C00105
1-Ethyl-3-[3-(pyrimidin-2-ylamino)- pyrido[2,3-b]pyrazin-6-yl]-urea,
257
Figure US08937068-20150120-C00106
6-[6-(3-Ethyl-ureido)-pyrido[2,3- b]pyrazin-3-ylamino]-naphthalin-2- carbonic acid,
258
Figure US08937068-20150120-C00107
1-Ethyl-3-[3-(4-hydroxy-quinolin-2- ylamino)-pyrido[2,3-b]pyrazin-6-yl]-urea,
259
Figure US08937068-20150120-C00108
1-Ethyl-3-[3-(quinolin-6-ylamino)- pyrido[2,3-b]pyrazin-6-yl]-urea,
260
Figure US08937068-20150120-C00109
1-Ethyl-3-[3-(3,4,5-trimethoxy- phenylamino)-pyrido[2,3-b]pyrazin-6-yl]- urea,
261
Figure US08937068-20150120-C00110
1-Ethyl-3-[3-(1H-indol-6-ylamino)- pyrido[2,3-b]pyrazin-6-yl]-urea,
263
Figure US08937068-20150120-C00111
1-Ethyl-3-[3-(quinoxalin-6-ylamino)- pyrido[2,3-b]pyrazin-6-yl]-urea,
264
Figure US08937068-20150120-C00112
1-Ethyl-3-[3-(3-trifluoromethoxy- phenylamino)-pyrido[2,3-b]pyrazin-6-yl]- urea,
265
Figure US08937068-20150120-C00113
1-Ethyl-3-[3-(4-isopropoxy- phenylamino)-pyrido[2,3-b]pyrazin-6-yl]- urea,
and 266
Figure US08937068-20150120-C00114
1-[3-(Dibenzofuran-2-ylamino)- pyrido[2,3-b]pyrazin-6-yl]-3-ethyl-urea.
6. The method according to claim 1, wherein the mammal is selected from the group consisting of a human, beef cattle, cow, sheep, pig, goat, horse, pony, donkey, hinny, mule, hare, rabbit, cat, dog, guinea pig, hamster, rat, and mouse.
7. The method according to claim 1, wherein said compound is administered as a pharmaceutical composition comprising a pharmacologically active quantity of said compound.
8. The method according to claim 1, wherein said compound is administered in a unit dose of 0.001 mg to 100 mg per kg body weight of a patient.
9. The method according to claim 1, wherein said compound is administered as a composition that contains at least one pharmaceutically compatible excipient and/or adjuvant.
10. The method of claim 1, wherein said mammal is human.
11. A method for reducing growth or proliferation of a mammalian tumor via inhibition of enzymes of the PI3K-Akt signal transduction pathway comprising administering to a mammal at least one compound of formula (I):
Figure US08937068-20150120-C00115
wherein the substituents R1, R2, R3, R4 have the following meanings:
R1 is NR7R8, wherein R7 and R8 independently of each other are hydrogen, alkyl, cycloalkyl, aryl, heteroaryl, alkyl-cycloalkyl, alkyl-heterocyclyl, alkyl-aryl or alkyl-heteroaryl; wherein said alkyl-, cycloalkyl-, aryl- and heteroaryl-, alkyl-cycloalkyl, alkyl-heterocyclyl, alkyl-aryl or alkyl-heteroaryl substituents are independent of each other optionally substituted by F, Cl, Br, I, CN, CF3, NH2, NH-alkyl, NH-aryl, N(alkyl)2, NO2, SH, S-alkyl, OH, OCF3, O-alkyl, O-aryl, OSO2H, OP(O)(OH)2, CHO, CO2H, SO3H or alkyl, and wherein R7 and R8 do not together form a heterocycle;
R2 is hydrogen,
R3 is —NR9R10, wherein R9 is hydrogen or alkyl and R10 is —C(Y)NR11R12, wherein Y═O or S and R11 and R12 are, independently,
(i) hydrogen,
(ii) unsubstituted or substituted alkyl, wherein the alkyl group is optionally substituted with one or more substitutents selected from the group consisting of F, Cl, Br, I, CF3, CN, NH2, NH-alkyl, NH-cycloalkyl, NH-heterocyclyl, NH-aryl, NH-heteroaryl, NH-alkyl-cycloalkyl, NH-alkyl-heterocyclyl, NH-alkyl-aryl, NH-alkyl-heteroaryl, N(alkyl)2, NHC(O)-alkyl, NHC(O)-cycloalkyl, NHC(O)-heterocyclyl, NHC(O)-aryl, NHC(O)-heteroaryl, NHC(O)-alkyl-aryl, NHC(O)-alkyl-heteroaryl, NHSO2-alkyl, NHSO2-cycloalkyl, NHSO2-heterocyclyl, NHSOr aryl, NHSO2-heteroaryl, NHSO2-alkyl-aryl, NHSO2-alkyl-heteroaryl, NO2, SH, S-alkyl, S-cycloalkyl, S-heterocyclyl, S-aryl, S-heteroaryl, OH, OCF3, O-alkyl, O-cycloalkyl, O-heterocyclyl, O-aryl, O-heteroaryl, O-alkyl-cycloalkyl, O-alkyl-heterocyclyl, O-alkyl-aryl, O-alkyl-heteroaryl, OC(O)-alkyl, OC(O)cycloalkyl, OC(O)-heterocyclyl, OC(O)-aryl, OC(O)-heteroaryl, OC(O)-alkyl-aryl, OC(O)alkyl-heteroaryl, OSO3H, OSO2-alkyl, OSO2-cycloalkyl, OSO2-heterocyclyl, OSO2-aryl, OSO2-heteroaryl, OSO2-alkyl-aryl, OSO2-alkyl-heteroaryl, OP(O)(OH)2, C(O)-alkyl, C(O)-aryl, C(O)-heteroaryl, CO2H, CO2-alkyl, CO2-cycloalkyl, CO2-heterocyclyl, CO2-aryl, CO2-heteroaryl, CO2-alkyl-cycloalkyl, CO2-alkyl-heterocyclyl, CO2-alkyl-aryl, CO2-alkyl-heteroaryl, C(O)—NH2, C(O)NH-alkyl, C(O)NH-cycloalkyl, C(O)NH-heterocyclyl, C(O)NH-aryl, C(O)NH-heteroaryl, C(O)NH-alkyl-cycloalkyl, C(O)NH-alkyl-heterocyclyl, C(O)NH-alkyl-aryl, C(O)NH-alkyl-heteroaryl, C(O)N(alkyl)2, C(O)N(cycloalkyl)2, C(O)N(aryl)2, C(O)N(heteroaryl)2, SO-alkyl, SO-aryl, SO2-alkyl, SO2-aryl, SO2NH2, SO2NH-alkyl, SO2NH-aryl, SO2NH-heteroaryl, SO2NH-alkyl-aryl, SO3H, SO2O-alkyl, SO2O-aryl, SO2O-alkyl-aryl, cycloalkyl, heterocyclyl, aryl and heteroaryl, which may be the same or different from each other;
(iii) unsubstituted or substituted cycloalkyl, wherein the cycloalkyl group is optionally substituted with one or more substituents selected from the group consisting of F, Cl, Br, I, NH2, NH-alkyl, NH-cycloalkyl, NH-heterocyclyl, NH-aryl, NH-heteroaryl, NH-alkyl-aryl, NH-alkyl-heteroaryl, N(alkyl)2, NHC(O)-alkyl, NHC(O)cycloalkyl, NHC(O)-heterocyclyl, NHC(O)-aryl, NHC(O)-heteroaryl, NHC(O)-alkyl-aryl, NHC(O)-alkyl-heteroaryl, NHSO2-alkyl, NHSO2-cycloalkyl, NHSO2-heterocyclyl, NHSO2-aryl, NHSO2-heteroaryl, NHSO2-alkyl-aryl, NHSO2-alkyl-heteroaryl, OH, O-alkyl, O-cycloalkyl, O-heterocyclyl, O-aryl, O-heteroaryl, O-alkyl-aryl, O-alkyl-heteroaryl, OC(O)alkyl, OC(O)-cycloalkyl, OC(O)-heterocyclyl, OC(O)-aryl, OC(O)-heteroaryl, OC(O)-alkyl-aryl, OC(O)-alkyl-heteroaryl, OSO3H, OSO2-alkyl, OSO2-cycloalkyl, OSO2-heterocyclyl, OSO2-aryl, OSO2-heteroaryl, OSO2-alkyl-aryl, OSO2-alkyl-heteroaryl, OP(O)(OH)2, CO2H, CO2-alkyl, CO2-cycloalkyl, CO2-heterocyclyl, CO2-aryl, CO2-heteroaryl, CO2-alkyl-cycloalkyl, CO2-alkyl-heterocyclyl, CO2-alkyl-aryl, CO2-alkyl-heteroaryl, C(O)—NH2, C(O)NH-alkyl, C(O)NH-cycloalkyl, C(O)NH-heterocyclyl, C(O)NH-aryl, C(O)NH-heteroaryl, C(O)NH-alkyl-cycloalkyl, C(O)NH-alkyl-heterocyclyl, C(O)NH-alkyl-aryl, C(O)NH-alkyl-heteroaryl, C(O)N(alkyl)2, C(O)N(cycloalkyl)2, C(O)N(aryl)2, C(O)N(heteroaryl)2, alkyl, and aryl, which may be the same or different from each other;
(iv) unsubstituted or substituted heterocyclyl, wherein the heterocyclyl group is optionally substituted with one or more substituents selected from the group consisting of OH, O-alkyl, O-aryl, NH2, NH-alkyl, NH-aryl, alkyl, alkyl-aryl and aryl, which may be the same or different from each other;
(v) unsubstituted or substituted aryl, wherein the aryl group is optionally substituted with one or more substituents selected from the group consisting of F, Cl, Br, I, CF3, CN, NH2, NH-alkyl, NH-cycloalkyl, NH-heterocyclyl, NH-aryl, NH-heteroaryl, NH-alkyl-cycloalkyl, NH-alkyl-heterocyclyl, NH-alkyl-aryl, NH-alkyl-heteroaryl, NH-alkyl NH2, NH-alkyl-OH, N(alkyl)2, NHC(O)-alkyl, NHC(O)-cycloalkyl, NHC(O)-heterocyclyl, NHC(O)-aryl, NHC(O)-heteroaryl, NHC(O)-alkyl-aryl, NHC(O)-alkyl-heteroaryl, NHSO2-alkyl, NHSO2-cycloalkyl, NHSO2-heterocyclyl, NHSO2-aryl, NHSO2-heteroaryl, NHSO2-alkyl-aryl, NHSO2-alkyl-heteroaryl, NO2, SH, S-alkyl, S-cycloalkyl, S-heterocyclyl, S-aryl, S-heteroaryl, OH, OCF3, O-alkyl, O-cycloalkyl, O-heterocyclyl, O-aryl, O-heteroaryl, O-alkyl-cycloalkyl, O-alkyl-heterocyclyl, O-alkyl-aryl, O-alkyl-heteroaryl, O-alkyl-OH, O(CH2)n—O, OC(O)-alkyl, OC(O)-cycloalkyl, OC(O)-heterocyclyl, OC(O)-aryl, OC(O)heteroaryl, OC(O)-alkyl-aryl, OC(O)-alkyl-heteroaryl, OSO3H, OSO2-alkyl, OSO2-cycloalkyl, OSO2-heterocyclyl, OSO2-aryl, OSO2-heteroaryl, OSO2-alkyl-aryl, OSO2-alkyl-heteroaryl, OP(O)(OH)2, C(O)-alkyl, C(O)-aryl, C(O)-heteroaryl, CO2H, CO2-alkyl, CO2-cycloalkyl, CO2-heterocyclyl, CO2-aryl, CO2-heteroaryl, CO2-alkyl-cycloalkyl, CO2-alkyl-heterocyclyl, CO2-alkyl-aryl, CO2-alkyl-heteroaryl, C(O)—NH2, C(O)NH-alkyl, C(O)NH-cycloalkyl, C(O)NH-heterocyclyl, C(O)NH-aryl, C(O)NH-heteroaryl, C(O)NH-alkyl-cycloalkyl, C(O)NH-alkyl-heterocyclyl, C(O)NH-alkyl-aryl, C(O)NH-alkyl-heteroaryl, C(O)N(alkyl)2, C(O)N(cycloalkyl)2, C(O)N(aryl)2, C(O)N(heteroaryl)2, SO-alkyl, SO-aryl, SO2-alkyl, SO2-aryl, SO2NH2, SO2NH-alkyl, SO2NH-aryl, SO2NH-heteroaryl, SO2NH-alkyl-aryl, SO3H, SO2O-alkyl, SO2O-aryl, SO2O-alkyl-aryl, alkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl, which may be the same or different from each other, and n can have the value 1, 2 or 3;
(vi) unsubstituted or substituted heteroaryl, wherein the heteroaryl group is optionally substituted with one or more substituents selected from the group consisting of F, Cl, Br, I, CF3, CN, NH2, NH-alkyl, NH-cycloalkyl, NH-heterocyclyl, NH-aryl, NH-heteroaryl, NH-alkyl-cycloalkyl, NH-alkyl-heterocyclyl, NH-alkyl-aryl, NH-alkyl-heteroaryl, NH-alkly-NH2, NH-alkyl-OH, N(alkyl)2, NHC(O)-alkyl, NHC(O)-cycloalkyl, NHC(O)-heterocyclyl, NHC(O)-aryl, NHC(O)-heteroaryl, NHC(O)-alkyl-aryl, NHC(O)-alkyl-heteroaryl, NHSO2-alkyl, NHSO2-cycloalkyl, NHSO2-heterocyclyl, NHSO2-aryl, NHSO2-heteroaryl, NHSO2-alkyl-aryl, NHSO2-alkyl-heteroaryl, NO2, SH, S-alkyl, S-aryl, S-heteroaryl, OH, OCF3, O-alkyl, O-cycloalkyl, O-aryl, O-heteroaryl, O-alkyl-cycloalkyl, O-alkyl-heterocyclyl, O-alkyl-aryl, O-alkyl-heteroaryl, OC(O)-alkyl, OC(O)-cycloalkyl, OC(O)-heterocyclyl, OC(O)-aryl, OC(O)-heteroaryl, OC(O)-alkyl-aryl, OC(O)-alkyl-heteroaryl, OSO3H, OSO2-alkyl, OSO2-cycloalkyl, OSO2-heterocyclyl, OSO2-aryl, OSO2-heteroaryl, OSO2-alkyl-aryl, OSO2-alkyl-heteroaryl, OP(O)(OH)2, C(O)-alkyl, C(O)-aryl, C(O)-heteroaryl, CO2H, CO2-alkyl, CO2-cycloalkyl, CO2-heterocyclyl, CO2-aryl, CO2-heteroaryl, CO2-alkyl-cycloalkyl, CO2-alkyl-heterocyclyl, CO2-alkyl-aryl, CO2-alkyl-heteroaryl, C(O)—NH2, C(O)NH-alkyl, C(O)NH-cycloalkyl, C(O)NH-heterocyclyl, C(O)NH-aryl, C(O)NH-heteroaryl, C(O)NH-alkyl-cycloalkyl, C(O)NH-alkyl-heterocyclyl, C(O)NH-alkyl-aryl, C(O)NH-alkyl-heteroaryl, C(O)N(alkyl)2, C(O)N(cycloalkyl)2, C(O)N(aryl)2, C(O)N(heteroaryl)2, SO2NH2, SO2NH-alkyl, SO2NH-aryl, SO2NH-heteroaryl, SO2NH-alkyl-aryl, SO3H, SO2O-alkyl, SO2O-aryl, SO2O-alkyl-aryl, alkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl, which may be the same or different from each other;
(vii) —C(O)—R17, wherein R17 is alkyl, aryl or heteroaryl, and the alkyl and aryl substituents are optionally substituted by F, Cl, Br, I, CN, CF3, NH2, NH-alkyl, NH-aryl, N(alkyl)2, NO2, SH, S-alkyl, OH, OCF3, O-alkyl, O-aryl, OSO2H, OP(O)(OH)2, CHO, CO2H, SO3H or alkyl;
(viii) or R11 and R12 together form heterocyclyl, and
R4 is hydrogen;
wherein said compound inhibits PI3K alpha, PI3K beta, PI3K gamma, and PI3K delta in the PI3K-Akt signal transduction pathway; and
wherein said malignant tumor is selected from the group consisting of a colon tumor, a gastric tumor, an intestinal tumor, a pulmonary tumor, a pancreatic tumor, an ovarian tumor, a prostatic tumor, melanoma, a hepatic tumor, a renal tumor, a head tumor, glioma, a breast tumor, cervico-uterine carcinoma, adeno-acanthoma, an endometrial cancer, a colorectal tumor, esophageal cancer, thyroid cancer, lymphoma, and leukemia.
12. The method of claim 11, wherein said compound is Compound 150:
Figure US08937068-20150120-C00116
13. The method of claim 11, wherein the administration of said compound increases apoptosis of said mammalian tumor.
14. The method according to claim 11, wherein R7, R8, R9, R11, or R12 is an alkyl group that is selected from the group consisting of methyl, ethyl, n-propyl, 2-propyl, n-butyl, sec-butyl, tert-butyl, n-pentyl, iso-pentyl, neo-pentyl, n-hexyl, 2-hexyl, n-octyl, ethylenyl (vinyl), ethynyl, propenyl, propynyl, butenyl, butynyl, pentenyl, pentynyl, hexenyl, hexynyl, heptenyl, heptynyl, octenyl and octynyl.
15. The method according to claim 11, wherein R7, R8, R11, or R12 is a heteroaryl group that is selected from the group consisting of pyrrolyl, furyl, thienyl, thiazolyl, oxazolyl, isoxazolyl, pyrazolyl, imidazolyl, pyridinyl, pyrimidinyl, pyridazinyl, pyrazinyl, phthalazinyl, indolyl, indazolyl, indolizinyl, quinolinyl, isoquinolinyl, quinoxalinyl, quinazolinyl, carbazolyl, phenazinyl, phenothiazinyl and acridinyl.
16. The method according to claim 11, wherein R11 or R12 is a heterocyclyl group or R11 and R12 together form heterocyclyl that is selected from the group consisting of tetrahydrofuryl, tetrahydropyranyl, pyrrolidinyl, piperidinyl, piperazinyl and morpholinyl.
17. The method according to claim 11, wherein said compound is selected from the group consisting of:
62
Figure US08937068-20150120-C00117
1-Ethyl-3-[3-(2-methoxy-ethylamino)- pyrido[2,3-b]pyrazin-6-yl]-urea,
65
Figure US08937068-20150120-C00118
1-[3-(Cyclopropylmethyl-amino)- pyrido[2,3-b]pyrazin-6-yl]-3-ethyl-urea,
66
Figure US08937068-20150120-C00119
1-Ethyl-3-{3-[(pyridin-4-ylmethyl)- amino]-pyrido[2,3-b]pyrazin-6-yl}-urea,
83
Figure US08937068-20150120-C00120
1-(3-Amino-pyrido[2,3-b]pyrazin-6-yl)-3- ethyl-urea,
150
Figure US08937068-20150120-C00121
1-Ethyl-3-(3-p-tolylamino-pyrido[2,3- b]pyrazin-6-yl)-urea,
157
Figure US08937068-20150120-C00122
1-Ethyl-3-(3-m-tolylamino-pyrido-[2,3- b]pyrazin-6-yl)-urea,
158
Figure US08937068-20150120-C00123
1-Ethyl-3-[3-(4-methoxy-phenylamino)- pyrido[2,3-b]pyrazin-6-yl]-urea,
159
Figure US08937068-20150120-C00124
1-[3-(4-Chloro-phenylamino)-pyrido- [2,3-b]pyrazin-6-yl]-3-ethyl-urea,
160
Figure US08937068-20150120-C00125
1-Ethyl-3-(3-o-tolylamino-pyrido[2,3-b]- pyrazin-6-yl)-urea,
161
Figure US08937068-20150120-C00126
1-Ethyl-3-[3-(pyridin-3-ylamino)-pyrido- [2,3-b]pyrazin-6-yl]-urea,
162
Figure US08937068-20150120-C00127
1-Ethyl-3-[3-(4-ethyl-phenylamino)- pyrido[2,3-b]pyrazin-6-yl]-urea,
163
Figure US08937068-20150120-C00128
1-Ethyl-3-[3-(3-methoxy-4-methyl- phenylamino)-pyrido[2,3-b]pyrazin-6-yl]- urea,
164
Figure US08937068-20150120-C00129
1-Ethyl-3-[3-(4-hydroxy-phenylamino)- pyrido[2,3-b]pyrazin-6-yl]-urea,
165
Figure US08937068-20150120-C00130
1-Ethyl-3-[3-(5-methyl-pyridin-2-yl- amino)-pyrido[2,3-b]pyrazin-6-yl]-urea,
166
Figure US08937068-20150120-C00131
1-Ethyl-3-[3-(1-methyl-1H-pyrazol-3- ylamino)-pyrido[2,3-b]pyrazin-6-yl]-urea,
167
Figure US08937068-20150120-C00132
1-Ethyl-3-[3-(4-fluoro-phenylamino)- pyrido[2,3-b]pyrazin-6-yl]-urea,
177
Figure US08937068-20150120-C00133
1-[3-(3-Chloro-4-methoxy-phenylamino)- pyrido[2,3-b]pyrazin-6-yl]-3-ethyl-urea,
178
Figure US08937068-20150120-C00134
1-Ethyl-3-[3-(naphthalin-2-ylamino)- pyrido[2,3-b]pyrazin-6-yl]-urea,
179
Figure US08937068-20150120-C00135
1-Ethyl-3-[3-(quinolin-3-ylamino)- pyrido[2,3-b]pyrazin-6-yl]-urea,
180
Figure US08937068-20150120-C00136
1-[3-(3,5-Dimethoxy-phenylamino)- pyrido[2,3-b]pyrazin-6-yl]-3-ethyl-urea,
181
Figure US08937068-20150120-C00137
1-Ethyl-3-[3-(pyrazin-2-ylamino)- pyrido[2,3-b]pyrazin-6-yl]-urea,
182
Figure US08937068-20150120-C00138
1-Ethyl-3-[3-(3-isopropoxy- phenylamino)-pyrido[2,3-b]pyrazin-6-yl]- urea,
184
Figure US08937068-20150120-C00139
1-[3-(2-Chloro-pyridin-4-ylamino)- pyrido[2,3-b]pyrazin-6-yl]-3-ethyl-urea,
185
Figure US08937068-20150120-C00140
1-[3-(3,5-Dichloro-4-hydroxy- phenylamino)-pyrido[2,3-b]pyrazin-6-yl]- 3-ethyl-urea,
187
Figure US08937068-20150120-C00141
1-[3-(3,4-Dimethoxy-phenylamino)- pyrido[2,3-b]pyrazin-6-yl]-3-ethyl-urea,
188
Figure US08937068-20150120-C00142
1-Ethyl-3-[3-(3-hydroxy-4-methyl- phenylamino)-pyrido[2,3-b]pyrazin-6-yl]- urea,
189
Figure US08937068-20150120-C00143
3-[6-(3-Ethyl-ureido)-pyrido[2,3-b]- pyrazin-3-ylamino]-5-trifluoromethyl- benzoic acid,
190
Figure US08937068-20150120-C00144
1-Ethyl-3-[3-(6-methoxy-pyridin-3- ylamino)-pyrido[2,3-b]pyrazin-6-yl]-urea,
191
Figure US08937068-20150120-C00145
1-[3-(3,5-Dimethyl-phenylamino)- pyrido[2,3-b]pyrazin-6-yl]-3-ethyl-urea,
192
Figure US08937068-20150120-C00146
1-[3-(4-Cyano-phenylamino)-pyrido-[2,3- b]pyrazin-6-yl]-3-ethyl-urea,
197
Figure US08937068-20150120-C00147
1-Ethyl-3-[3-(4-hydroxy-3-methoxy- phenylamino)-pyrido[2,3-b]pyrazin-6-yl]- urea,
211
Figure US08937068-20150120-C00148
1-Ethyl-3-[3-(methyl-p-tolyl-amino)- pyrido[2,3-b]pyrazin-6-yl]-urea,
212
Figure US08937068-20150120-C00149
1-Ethyl-3-[3-(2-p-tolyl-ethylamino)- pyrido[2,3-b]pyrazin-6-yl]-urea,
213
Figure US08937068-20150120-C00150
1-Ethyl-3-[3-(4-methyl-benzylamino)- pyrido[2,3-b]pyrazin-6-yl]-urea,
214
Figure US08937068-20150120-C00151
1-Ethyl-3-[3-(3-fluoro-4-methyl-phenyl- amino)-pyrido[2,3-b]pyrazin-6-yl]-urea,
215
Figure US08937068-20150120-C00152
1-[3-(3,4-Dimethyl-phenylamino)- pyrido[2,3-b]pyrazin-6-yl]-3-ethyl-urea,
216
Figure US08937068-20150120-C00153
1-Ethyl-3-[3-(4-isopropyl-phenylamino)- pyrido[2,3-b]pyrazin-6-yl]-urea,
240
Figure US08937068-20150120-C00154
1-[3-(Benzothiazol-6-ylamino)- pyrido[2,3-b]pyrazin-6-yl]-3-ethyl-urea,
241
Figure US08937068-20150120-C00155
1-Ethyl-3-[3-(4-methyl-3- trifluoroomethyl-phenylamino)- pyrido[2,3-b]pyrazin-6-yl]-urea,
242
Figure US08937068-20150120-C00156
1-[3-(3-Cyano-4-methyl-phenylamino)- pyrido[2,3-b]pyrazin-6-yl]-3-ethyl-urea,
243
Figure US08937068-20150120-C00157
1-Ethyl-3-[3-(4-phenoxy-phenylamino)- pyrido[2,3-b]pyrazin-6-yl]-urea,
244
Figure US08937068-20150120-C00158
1-[3-(4-Chloro-3-methyl-phenylamino)- pyrido[2,3-b]pyrazin-6-yl]-3-ethyl-urea,
245
Figure US08937068-20150120-C00159
1-[3-(2-Chloro-4-methyl-phenylamino)- pyrido[2,3-b]pyrazin-6-yl]-3-ethyl-urea,
246
Figure US08937068-20150120-C00160
1-Ethyl-3-[3-(3-trifluoromethyl- phenylamino)-pyrido[2,3-b]pyrazin-6-yl]- urea,
247
Figure US08937068-20150120-C00161
1-[3-(2-Chloro-4-trifluoromethoxy- phenylamino)-pyrido[2,3-b]pyrazin-6-yl]- 3-ethyl-urea,
248
Figure US08937068-20150120-C00162
1-[3-(4-Chloro-2-methoxy-5-methyl- phenylamino)-pyrido[2,3-b]pyrazin-6-yl]- 3-ethyl-urea,
252
Figure US08937068-20150120-C00163
1-[3-(Benzo[1,3]-dioxol-5-ylamino)- pyrido[2,3-b]pyrazin-6-yl]-3-ethyl-urea,
253
Figure US08937068-20150120-C00164
3-[6-(3-Ethyl-ureido)-pyrido[2,3- b]pyrazin-3-ylamino]-benzoic acid,
254
Figure US08937068-20150120-C00165
2-Chloro-4-[6-(3-ethyl-ureido)- pyrido[2,3-b]pyrazin-3-ylamino]-benzoic acid,
255
Figure US08937068-20150120-C00166
1-Ethyl-3-[3-(3-methoxy-5- trifluoromethyl-phenylamino)-pyrido[2,3- b]pyrazin-6-yl]-urea,
256
Figure US08937068-20150120-C00167
1-Ethyl-3-[3-(pyrimidin-2-ylamino)- pyrido[2,3-b]pyrazin-6-yl]-urea,
257
Figure US08937068-20150120-C00168
6-[6-(3-Ethyl-ureido)-pyrido[2,3- b]pyrazin-3-ylamino]-naphthalin-2- carbonic acid,
258
Figure US08937068-20150120-C00169
1-Ethyl-3-[3-(4-hydroxy-quinolin-2- ylamino)-pyrido[2,3-b]pyrazin-6-yl]-urea,
259
Figure US08937068-20150120-C00170
1-Ethyl-3-[3-(quinolin-6-ylamino)- pyrido[2,3-b]pyrazin-6-yl]-urea,
260
Figure US08937068-20150120-C00171
1-Ethyl-3-[3-(3,4,5-trimethoxy- phenylamino)-pyrido[2,3-b]pyrazin-6-yl]- urea,
261
Figure US08937068-20150120-C00172
1-Ethyl-3-[3-(1H-indol-6-ylamino)- pyrido[2,3-b]pyrazin-6-yl]-urea,
263
Figure US08937068-20150120-C00173
1-Ethyl-3-[3-(quinoxalin-6-ylamino)- pyrido[2,3-b]pyrazin-6-yl]-urea,
264
Figure US08937068-20150120-C00174
1-Ethyl-3-[3-(3-trifluoromethoxy- phenylamino)-pyrido[2,3-b]pyrazin-6-yl]- urea,
265
Figure US08937068-20150120-C00175
1-Ethyl-3-[3-(4-isopropoxy- phenylamino)-pyrido[2,3-b]pyrazin-6-yl]- urea,
and 266
Figure US08937068-20150120-C00176
1-[3-(Dibenzofuran-2-ylamino)- pyrido[2,3-b]pyrazin-6-yl]-3-ethyl-urea.
18. The method according to claim 11, wherein the mammal is selected from the group consisting of a human, beef cattle, cow, sheep, pig, goat, horse, pony, donkey, hinny, mule, hare, rabbit, cat, dog, guinea pig, hamster, rat, and mouse.
19. The method according to claim 11, wherein said compound is administered as a pharmaceutical composition comprising a pharmacologically active quantity of said compound.
20. The method according to claim 11, wherein said compound is administered in a unit dose of 0.001 mg to 100 mg per kg body weight of a patient.
21. The method according to claim 11, wherein said compound is administered as a composition that contains at least one pharmaceutically compatible excipient and/or adjuvant.
22. The method of claim 11, wherein said mammal is human.
US11/558,493 2005-11-11 2006-11-10 Pyridopyrazine derivatives and their use Expired - Fee Related US8937068B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/558,493 US8937068B2 (en) 2005-11-11 2006-11-10 Pyridopyrazine derivatives and their use

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US73570705P 2005-11-11 2005-11-11
EP05024692A EP1790342A1 (en) 2005-11-11 2005-11-11 Pyridopyrazine derivatives and their use as signal transduction modulators
US11/558,493 US8937068B2 (en) 2005-11-11 2006-11-10 Pyridopyrazine derivatives and their use

Publications (2)

Publication Number Publication Date
US20070123494A1 US20070123494A1 (en) 2007-05-31
US8937068B2 true US8937068B2 (en) 2015-01-20

Family

ID=36424666

Family Applications (1)

Application Number Title Priority Date Filing Date
US11/558,493 Expired - Fee Related US8937068B2 (en) 2005-11-11 2006-11-10 Pyridopyrazine derivatives and their use

Country Status (21)

Country Link
US (1) US8937068B2 (en)
EP (2) EP1790342A1 (en)
JP (1) JP5448450B2 (en)
KR (1) KR101473662B1 (en)
CN (1) CN101330918B (en)
AR (1) AR056216A1 (en)
AU (1) AU2006334721B2 (en)
BR (1) BRPI0618451A2 (en)
CA (1) CA2628186A1 (en)
ES (1) ES2543962T3 (en)
HK (1) HK1125823A1 (en)
IL (1) IL191160A (en)
NO (1) NO20082509L (en)
NZ (1) NZ595879A (en)
PL (1) PL1962854T3 (en)
RU (1) RU2487713C2 (en)
SG (1) SG10201401061XA (en)
TW (1) TWI423973B (en)
UA (1) UA95464C2 (en)
WO (1) WO2007079999A2 (en)
ZA (1) ZA200803867B (en)

Families Citing this family (35)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090118139A1 (en) * 2000-11-07 2009-05-07 Caliper Life Sciences, Inc. Microfluidic method and system for enzyme inhibition activity screening
AU2006313701B2 (en) * 2005-11-11 2012-05-31 Aeterna Zentaris Gmbh Novel pyridopyrazines and their use as modulators of kinases
US9139630B2 (en) 2006-07-11 2015-09-22 Rutgers, The State University Of New Jersey Compositions and methods for preparing recombinant MG53 and methods for optimizing same
US7893066B2 (en) * 2006-10-05 2011-02-22 Gilead Palo Alto, Inc. Pyridol[2,3-B]pyrazinones for use as stearoyl CoA desaturase inhibitors
EP1990342A1 (en) 2007-05-10 2008-11-12 AEterna Zentaris GmbH Pyridopyrazine Derivatives, Process of Manufacturing and Uses thereof
CN101932609B (en) * 2007-12-04 2016-03-23 罗格斯新泽西州立大学 The composition heavily sealed in order to regulate cytolemma and method
US8399483B2 (en) 2007-12-21 2013-03-19 Ucb Pharma S.A. Quinoxaline and quinoline derivatives as kinase inhibitors
US20100267752A1 (en) * 2008-10-15 2010-10-21 Gilead Palo Alto, Inc. 3-HYDROQUINAZOLIN-4-ONE DERIVATIVES FOR USE AS STEAROYL CoA DESATURASE INHIBITORS
WO2010052448A2 (en) * 2008-11-05 2010-05-14 Ucb Pharma S.A. Fused pyrazine derivatives as kinase inhibitors
GB201007286D0 (en) 2010-04-30 2010-06-16 Astex Therapeutics Ltd New compounds
US20130123340A1 (en) * 2010-05-11 2013-05-16 Peking University, Office Of Technology Transfer Compositions and methods for the treatment and prevention of cardiac ischemic injury
KR101496794B1 (en) 2010-11-05 2015-02-27 유니버시티 오브 메디신 앤드 덴티스트리 오브 뉴 저지 Serum mg53 as a diagnostic marker for tissue injury
GB201020179D0 (en) 2010-11-29 2011-01-12 Astex Therapeutics Ltd New compounds
EP2508184A1 (en) * 2011-04-06 2012-10-10 Æterna Zentaris GmbH Pyridopyrazine derivatives and their use
EP2727920B1 (en) 2011-07-29 2016-11-02 FUJIFILM Corporation 1,5-naphthyridine derivative or salt thereof
GB201118652D0 (en) 2011-10-28 2011-12-07 Astex Therapeutics Ltd New compounds
GB201118654D0 (en) 2011-10-28 2011-12-07 Astex Therapeutics Ltd New compounds
GB201118656D0 (en) 2011-10-28 2011-12-07 Astex Therapeutics Ltd New compounds
GB201118675D0 (en) 2011-10-28 2011-12-14 Astex Therapeutics Ltd New compounds
GB201209613D0 (en) 2012-05-30 2012-07-11 Astex Therapeutics Ltd New compounds
GB201209609D0 (en) 2012-05-30 2012-07-11 Astex Therapeutics Ltd New compounds
WO2014109414A1 (en) * 2013-01-11 2014-07-17 富士フイルム株式会社 Nitrogen-containing heterocylic compound or salt thereof
GB201307577D0 (en) 2013-04-26 2013-06-12 Astex Therapeutics Ltd New compounds
HUE053654T2 (en) 2014-03-26 2021-07-28 Astex Therapeutics Ltd Combinations of fgfr- and cmet-inhibitors for the treatment of cancer
JO3512B1 (en) 2014-03-26 2020-07-05 Astex Therapeutics Ltd Quinoxaline derivatives useful as fgfr kinase modulators
RU2715893C2 (en) 2014-03-26 2020-03-04 Астекс Терапьютикс Лтд Combination of inhibitor fgfr and inhibitor igf1r
JOP20200201A1 (en) 2015-02-10 2017-06-16 Astex Therapeutics Ltd Pharmaceutical compositions comprising n-(3,5-dimethoxyphenyl)-n'-(1-methylethyl)-n-[3-(1-methyl-1h-pyrazol-4-yl)quinoxalin-6-yl]ethane-1,2-diamine
US10478494B2 (en) 2015-04-03 2019-11-19 Astex Therapeutics Ltd FGFR/PD-1 combination therapy for the treatment of cancer
WO2016203335A1 (en) * 2015-06-18 2016-12-22 Pfizer Inc. Novel pyrido[2,3-b]pyrazinones as bet-family bromodomain inhibitors
SI3353177T1 (en) 2015-09-23 2020-08-31 Janssen Pharmaceutica Nv Tricyclic heterocycles for the treatment of cancer
EP3353164B1 (en) 2015-09-23 2021-11-03 Janssen Pharmaceutica, N.V. Bi-heteroaryl substituted 1,4-benzodiazepines and uses thereof for the treatment of cancer
CN107619388A (en) * 2016-07-13 2018-01-23 南京天印健华医药科技有限公司 Heterocyclic compound as FGFR inhibitor
WO2018237190A1 (en) 2017-06-22 2018-12-27 City Of Hope Pyridopyrazine compounds and uses thereof
US20210238605A1 (en) * 2018-06-15 2021-08-05 INSERM (Institut National de la Santé et de la Recherche Médicale) Use of pi3kc2b inhibitors for the preservation of vascular endothelial cell barrier integrity
CN111892597B (en) * 2020-09-01 2023-07-25 山西天宏达安医药科技有限公司 Quinoxalinyl pyridopyrazine compound and preparation method and application thereof

Citations (69)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3180868A (en) 1963-07-24 1965-04-27 American Home Prod 3, 6-diamino-n-(substituted)pyrido[2, 3-b]-pyrazine-2-carboxamides
US3209004A (en) 1963-05-17 1965-09-28 American Home Prod 3, 6-diamino-n-(2, 2-dialkoxyethyl) pyrido[2, 3-b] pyrazine-2-carboxamides
GB1184848A (en) 1967-04-19 1970-03-18 Degussa A Process for the Production of 2,6-Dichloro-3-Nitropyridine
JPS5053394A (en) 1973-09-20 1975-05-12
US4082845A (en) 1977-04-25 1978-04-04 Merck & Co., Inc. 3-(1-Piperazinyl)-pyrido[2,3-b]pyrazines
WO1995015758A1 (en) 1993-12-10 1995-06-15 Rhone-Poulenc Rorer Pharmaceuticals Inc. Aryl and heteroaryl quinazoline compounds which inhibit csf-1r receptor tyrosine kinase
US5480883A (en) 1991-05-10 1996-01-02 Rhone-Poulenc Rorer Pharmaceuticals Inc. Bis mono- and bicyclic aryl and heteroaryl compounds which inhibit EGF and/or PDGF receptor tyrosine kinase
EP0735025B1 (en) 1995-03-28 1998-07-01 Sumitomo Chemical Company Limited Process for producing aminonitropyridines
WO1999017759A2 (en) 1997-10-06 1999-04-15 Asta Medica Aktiengesellschaft Methods of modulating serine/threonine protein kinase function with 5-azaquinoxaline-based compounds
WO1999032111A1 (en) 1997-12-22 1999-07-01 Bayer Corporation INHIBITION OF p38 KINASE ACTIVITY USING SUBSTITUTED HETEROCYCLIC UREAS
WO1999043681A1 (en) 1998-02-26 1999-09-02 Neurogen Corporation 4-(4-piperidylmethylamino) substituted heteroaryl fused pyridines: gaba brain receptor ligands
WO2000012497A2 (en) 1998-08-28 2000-03-09 Scios Inc. Quinazoline derivatives as medicaments
WO2000035435A1 (en) 1998-12-15 2000-06-22 Warner-Lambert Company Use of a mek inhibitor for preventing transplant rejection
WO2000037141A1 (en) 1998-12-22 2000-06-29 Warner-Lambert Company Combination chemotherapy
WO2001081346A2 (en) 2000-04-25 2001-11-01 Icos Corporation Inhibitors of human phosphatidyl-inositol 3-kinase delta
WO2002006213A2 (en) 2000-07-19 2002-01-24 Warner-Lambert Company Oxygenated esters of 4-iodo phenylamino benzhydroxamic acids
WO2002055079A2 (en) 2000-10-12 2002-07-18 Merck & Co Inc Aza-and polyaza-naphthalenyl carboxamides useful as hiv integrase inhibitors
WO2002090355A1 (en) 2001-05-05 2002-11-14 Smithkline Beecham P.L.C. N-aroyl cyclic amines
WO2003024448A2 (en) 2001-09-14 2003-03-27 Methylgene, Inc. Inhibitors of histone deacetylase
WO2003064421A1 (en) 2002-01-29 2003-08-07 Glaxo Group Limited Aminopiperidine derivatives
WO2003064431A2 (en) 2002-01-29 2003-08-07 Glaxo Group Limited Aminopiperidine compounds, process for their preparation, and pharmaceutical compositions containing them
WO2003068223A1 (en) 2002-02-11 2003-08-21 Bayer Corporation Aryl ureas with raf kinase and angiogenesis inhibiting activity
WO2003084473A2 (en) 2002-04-08 2003-10-16 Merck & Co., Inc. Method of treating cancer
WO2003086403A1 (en) 2002-04-08 2003-10-23 Merck & Co., Inc. Inhibitors of akt activity
WO2003086394A1 (en) 2002-04-08 2003-10-23 Merck & Co., Inc. Inhibitors of akt activity
WO2004005472A2 (en) 2002-07-02 2004-01-15 Southern Research Institute Inhibitors of ftsz and uses thereof
WO2004017950A2 (en) 2002-08-22 2004-03-04 Piramed Limited Phosphadidylinositol 3,5-biphosphate inhibitors as anti-viral agents
WO2004030635A2 (en) 2002-10-03 2004-04-15 Targegen, Inc. Vasculostatic agents and methods of use thereof
US20040092521A1 (en) 2002-11-12 2004-05-13 Altenbach Robert J. Bicyclic-substituted amines as histamine-3 receptor ligands
US20040102360A1 (en) 2002-10-30 2004-05-27 Barnett Stanley F. Combination therapy
WO2004055003A1 (en) 2002-12-13 2004-07-01 Neurogen Corporation 2-substituted quinazolin-4-ylamine analogues as capsaicin receptor modulators
WO2004104003A1 (en) 2003-05-23 2004-12-02 Zentaris Gmbh Novel pyridopyrazines and use thereof as kinase modulators
WO2004104002A1 (en) 2003-05-23 2004-12-02 Zentaris Gmbh Novel pyridopyrazines and use thereof as kinase inhibitors
WO2004108702A1 (en) 2003-06-05 2004-12-16 Zentaris Gmbh Indole derivatives with apoptosis-inducing effect
WO2005007099A2 (en) 2003-07-10 2005-01-27 Imclone Systems Incorporated Pkb inhibitors as anti-tumor agents
WO2005021513A1 (en) 2003-08-27 2005-03-10 Astrazeneca Ab Novel condensed n-pyrazinyl-sulphonamides and their use in the treatment of chemokine mediated diseases
WO2005023771A1 (en) 2003-09-05 2005-03-17 Ono Pharmaceutical Co., Ltd. Chemokine receptor antagonist and medical use thereof
WO2005023807A2 (en) 2003-09-09 2005-03-17 Neurogen Corporation 4 - heterobicyclyamino - substituted quinazolines and analogues therof as capsaicin - antagonists
WO2005056547A2 (en) 2003-12-04 2005-06-23 Vertex Pharmaceuticals Incorporated Quinoxalines useful as inhibitors of protein kinases
WO2005061519A1 (en) 2003-12-19 2005-07-07 Takeda San Diego, Inc. Kinase inhibitors
US20050165028A1 (en) 2004-01-23 2005-07-28 Norman Mark H. Vanilloid receptor ligands and their use in treatments
WO2005103029A1 (en) 2004-04-14 2005-11-03 Boehringer Ingelheim International Gmbh Novel alkyne compounds having an mch-antagonistic effect and medicaments containing said compounds
US20050256309A1 (en) 2004-05-12 2005-11-17 Altenbach Robert J Tri-and bi-cyclic heteroaryl histamine-3 receptor ligands
US20050256118A1 (en) 2004-05-12 2005-11-17 Altenbach Robert J Bicyclic-substituted amines having cyclic-substituted monocyclic substituents
US20050272736A1 (en) 2004-05-12 2005-12-08 Altenbach Robert J Tri- and bi-cyclic heteroaryl histamine-3 receptor ligands
US20050272728A1 (en) 2004-05-12 2005-12-08 Altenbach Robert J Bicyclic-substituted amines having cyclic-substituted monocyclic substituents
WO2005123733A1 (en) 2004-06-22 2005-12-29 Syngenta Participations Ag Pyridopyrazines for combatting phytopathogenic fungi
WO2005123698A1 (en) 2004-06-22 2005-12-29 Syngenta Participations Ag Fungicides based on nitrogen-containing heterocycles
WO2006002047A2 (en) 2004-06-15 2006-01-05 Glaxo Group Limited Antibacterial agents
WO2006012396A1 (en) 2004-07-22 2006-02-02 Glaxo Group Limited Antibacterial agents
WO2006014580A1 (en) 2004-07-08 2006-02-09 Glaxo Group Limited Antibacterial agents
WO2006017326A1 (en) 2004-07-13 2006-02-16 Glaxo Group Limited Antibacterial agents
WO2006017468A2 (en) 2004-08-02 2006-02-16 Glaxo Group Limited Antibacterial agents
WO2006020561A2 (en) 2004-08-09 2006-02-23 Glaxo Group Limited Antibacterial agents
WO2006021448A1 (en) 2004-08-25 2006-03-02 Morphochem Aktiengesellschaft für kombinatorische Chemie Novel compounds having an anti-bacterial activity
WO2006024666A1 (en) 2004-09-03 2006-03-09 Applied Research Systems Ars Holding N.V. Pyridine methylene azolidinones and use thereof phosphoinositide inhibitors
JP2006137723A (en) 2004-11-15 2006-06-01 Kyowa Hakko Kogyo Co Ltd Sulfonamide derivative
WO2006059103A2 (en) 2004-12-03 2006-06-08 Peakdale Molecular Limited Pyridine based compounds useful as intermediates for pharmaceutical or agricultural end-products
WO2006074147A2 (en) 2005-01-03 2006-07-13 Myriad Genetics, Inc. Nitrogen containing bicyclic compounds and therapeutical use thereof
WO2006081264A1 (en) 2005-01-25 2006-08-03 Glaxo Group Limited Antibacterial agents
WO2006081182A2 (en) 2005-01-25 2006-08-03 Glaxo Group Limited Antibacterial agents
WO2006081179A1 (en) 2005-01-25 2006-08-03 Glaxo Group Limited Antibacterial agents
WO2006081178A2 (en) 2005-01-25 2006-08-03 Glaxo Group Limited Antibacterial agents
WO2006091395A2 (en) 2005-02-14 2006-08-31 Merck & Co., Inc. Inhibitors of akt activity
WO2006128172A2 (en) 2005-05-26 2006-11-30 Synta Pharmaceuticals Corp. Method for treating b cell regulated autoimmune disorders
WO2006128129A2 (en) 2005-05-26 2006-11-30 Synta Pharmaceuticals Corp. Method for treating cancer
WO2007023186A1 (en) 2005-08-26 2007-03-01 Laboratoires Serono S.A. Pyrazine derivatives and use as pi3k inhibitors
WO2007044729A2 (en) 2005-10-07 2007-04-19 Exelixis, Inc. N- (3-amino-quinoxalin-2-yl) -sulfonamide derivatives and their use as phosphatidylinositol 3-kinase inhibitors
WO2007054556A1 (en) 2005-11-11 2007-05-18 Æterna Zentaris Gmbh Novel pyridopyrazines and their use as modulators of kinases

Patent Citations (72)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3209004A (en) 1963-05-17 1965-09-28 American Home Prod 3, 6-diamino-n-(2, 2-dialkoxyethyl) pyrido[2, 3-b] pyrazine-2-carboxamides
US3180868A (en) 1963-07-24 1965-04-27 American Home Prod 3, 6-diamino-n-(substituted)pyrido[2, 3-b]-pyrazine-2-carboxamides
GB1184848A (en) 1967-04-19 1970-03-18 Degussa A Process for the Production of 2,6-Dichloro-3-Nitropyridine
JPS5053394A (en) 1973-09-20 1975-05-12
US4082845A (en) 1977-04-25 1978-04-04 Merck & Co., Inc. 3-(1-Piperazinyl)-pyrido[2,3-b]pyrazines
US5480883A (en) 1991-05-10 1996-01-02 Rhone-Poulenc Rorer Pharmaceuticals Inc. Bis mono- and bicyclic aryl and heteroaryl compounds which inhibit EGF and/or PDGF receptor tyrosine kinase
WO1995015758A1 (en) 1993-12-10 1995-06-15 Rhone-Poulenc Rorer Pharmaceuticals Inc. Aryl and heteroaryl quinazoline compounds which inhibit csf-1r receptor tyrosine kinase
EP0735025B1 (en) 1995-03-28 1998-07-01 Sumitomo Chemical Company Limited Process for producing aminonitropyridines
WO1999017759A2 (en) 1997-10-06 1999-04-15 Asta Medica Aktiengesellschaft Methods of modulating serine/threonine protein kinase function with 5-azaquinoxaline-based compounds
WO1999032111A1 (en) 1997-12-22 1999-07-01 Bayer Corporation INHIBITION OF p38 KINASE ACTIVITY USING SUBSTITUTED HETEROCYCLIC UREAS
WO1999043681A1 (en) 1998-02-26 1999-09-02 Neurogen Corporation 4-(4-piperidylmethylamino) substituted heteroaryl fused pyridines: gaba brain receptor ligands
WO2000012497A2 (en) 1998-08-28 2000-03-09 Scios Inc. Quinazoline derivatives as medicaments
WO2000035435A1 (en) 1998-12-15 2000-06-22 Warner-Lambert Company Use of a mek inhibitor for preventing transplant rejection
WO2000037141A1 (en) 1998-12-22 2000-06-29 Warner-Lambert Company Combination chemotherapy
WO2001081346A2 (en) 2000-04-25 2001-11-01 Icos Corporation Inhibitors of human phosphatidyl-inositol 3-kinase delta
WO2002006213A2 (en) 2000-07-19 2002-01-24 Warner-Lambert Company Oxygenated esters of 4-iodo phenylamino benzhydroxamic acids
WO2002055079A2 (en) 2000-10-12 2002-07-18 Merck & Co Inc Aza-and polyaza-naphthalenyl carboxamides useful as hiv integrase inhibitors
WO2002090355A1 (en) 2001-05-05 2002-11-14 Smithkline Beecham P.L.C. N-aroyl cyclic amines
WO2003024448A2 (en) 2001-09-14 2003-03-27 Methylgene, Inc. Inhibitors of histone deacetylase
WO2003064421A1 (en) 2002-01-29 2003-08-07 Glaxo Group Limited Aminopiperidine derivatives
WO2003064431A2 (en) 2002-01-29 2003-08-07 Glaxo Group Limited Aminopiperidine compounds, process for their preparation, and pharmaceutical compositions containing them
WO2003068223A1 (en) 2002-02-11 2003-08-21 Bayer Corporation Aryl ureas with raf kinase and angiogenesis inhibiting activity
WO2003084473A2 (en) 2002-04-08 2003-10-16 Merck & Co., Inc. Method of treating cancer
WO2003086403A1 (en) 2002-04-08 2003-10-23 Merck & Co., Inc. Inhibitors of akt activity
WO2003086394A1 (en) 2002-04-08 2003-10-23 Merck & Co., Inc. Inhibitors of akt activity
WO2004005472A2 (en) 2002-07-02 2004-01-15 Southern Research Institute Inhibitors of ftsz and uses thereof
WO2004017950A2 (en) 2002-08-22 2004-03-04 Piramed Limited Phosphadidylinositol 3,5-biphosphate inhibitors as anti-viral agents
WO2004030635A2 (en) 2002-10-03 2004-04-15 Targegen, Inc. Vasculostatic agents and methods of use thereof
US20040102360A1 (en) 2002-10-30 2004-05-27 Barnett Stanley F. Combination therapy
US20040092521A1 (en) 2002-11-12 2004-05-13 Altenbach Robert J. Bicyclic-substituted amines as histamine-3 receptor ligands
WO2004055003A1 (en) 2002-12-13 2004-07-01 Neurogen Corporation 2-substituted quinazolin-4-ylamine analogues as capsaicin receptor modulators
WO2004104003A1 (en) 2003-05-23 2004-12-02 Zentaris Gmbh Novel pyridopyrazines and use thereof as kinase modulators
WO2004104002A1 (en) 2003-05-23 2004-12-02 Zentaris Gmbh Novel pyridopyrazines and use thereof as kinase inhibitors
US20040266777A1 (en) 2003-05-23 2004-12-30 Eckhard Claus Pyridopyrazines and the use thereof as kinase inhibitors
US20050032803A1 (en) 2003-05-23 2005-02-10 Eckhard Claus Pyridopyrazines and the use thereof as kinase inhibitors
WO2004108702A1 (en) 2003-06-05 2004-12-16 Zentaris Gmbh Indole derivatives with apoptosis-inducing effect
WO2005007099A2 (en) 2003-07-10 2005-01-27 Imclone Systems Incorporated Pkb inhibitors as anti-tumor agents
WO2005021513A1 (en) 2003-08-27 2005-03-10 Astrazeneca Ab Novel condensed n-pyrazinyl-sulphonamides and their use in the treatment of chemokine mediated diseases
WO2005023771A1 (en) 2003-09-05 2005-03-17 Ono Pharmaceutical Co., Ltd. Chemokine receptor antagonist and medical use thereof
EP1661889A1 (en) 2003-09-05 2006-05-31 Ono Pharmaceutical Co., Ltd. Chemokine receptor antagonist and medical use thereof
WO2005023807A2 (en) 2003-09-09 2005-03-17 Neurogen Corporation 4 - heterobicyclyamino - substituted quinazolines and analogues therof as capsaicin - antagonists
WO2005056547A2 (en) 2003-12-04 2005-06-23 Vertex Pharmaceuticals Incorporated Quinoxalines useful as inhibitors of protein kinases
WO2005061519A1 (en) 2003-12-19 2005-07-07 Takeda San Diego, Inc. Kinase inhibitors
US20050165028A1 (en) 2004-01-23 2005-07-28 Norman Mark H. Vanilloid receptor ligands and their use in treatments
WO2005103029A1 (en) 2004-04-14 2005-11-03 Boehringer Ingelheim International Gmbh Novel alkyne compounds having an mch-antagonistic effect and medicaments containing said compounds
US20050256118A1 (en) 2004-05-12 2005-11-17 Altenbach Robert J Bicyclic-substituted amines having cyclic-substituted monocyclic substituents
US20050272736A1 (en) 2004-05-12 2005-12-08 Altenbach Robert J Tri- and bi-cyclic heteroaryl histamine-3 receptor ligands
US20050272728A1 (en) 2004-05-12 2005-12-08 Altenbach Robert J Bicyclic-substituted amines having cyclic-substituted monocyclic substituents
US20050256309A1 (en) 2004-05-12 2005-11-17 Altenbach Robert J Tri-and bi-cyclic heteroaryl histamine-3 receptor ligands
WO2006002047A2 (en) 2004-06-15 2006-01-05 Glaxo Group Limited Antibacterial agents
WO2005123733A1 (en) 2004-06-22 2005-12-29 Syngenta Participations Ag Pyridopyrazines for combatting phytopathogenic fungi
WO2005123698A1 (en) 2004-06-22 2005-12-29 Syngenta Participations Ag Fungicides based on nitrogen-containing heterocycles
WO2006014580A1 (en) 2004-07-08 2006-02-09 Glaxo Group Limited Antibacterial agents
WO2006017326A1 (en) 2004-07-13 2006-02-16 Glaxo Group Limited Antibacterial agents
WO2006012396A1 (en) 2004-07-22 2006-02-02 Glaxo Group Limited Antibacterial agents
WO2006017468A2 (en) 2004-08-02 2006-02-16 Glaxo Group Limited Antibacterial agents
WO2006020561A2 (en) 2004-08-09 2006-02-23 Glaxo Group Limited Antibacterial agents
WO2006021448A1 (en) 2004-08-25 2006-03-02 Morphochem Aktiengesellschaft für kombinatorische Chemie Novel compounds having an anti-bacterial activity
WO2006024666A1 (en) 2004-09-03 2006-03-09 Applied Research Systems Ars Holding N.V. Pyridine methylene azolidinones and use thereof phosphoinositide inhibitors
JP2006137723A (en) 2004-11-15 2006-06-01 Kyowa Hakko Kogyo Co Ltd Sulfonamide derivative
WO2006059103A2 (en) 2004-12-03 2006-06-08 Peakdale Molecular Limited Pyridine based compounds useful as intermediates for pharmaceutical or agricultural end-products
WO2006074147A2 (en) 2005-01-03 2006-07-13 Myriad Genetics, Inc. Nitrogen containing bicyclic compounds and therapeutical use thereof
WO2006081264A1 (en) 2005-01-25 2006-08-03 Glaxo Group Limited Antibacterial agents
WO2006081182A2 (en) 2005-01-25 2006-08-03 Glaxo Group Limited Antibacterial agents
WO2006081179A1 (en) 2005-01-25 2006-08-03 Glaxo Group Limited Antibacterial agents
WO2006081178A2 (en) 2005-01-25 2006-08-03 Glaxo Group Limited Antibacterial agents
WO2006091395A2 (en) 2005-02-14 2006-08-31 Merck & Co., Inc. Inhibitors of akt activity
WO2006128172A2 (en) 2005-05-26 2006-11-30 Synta Pharmaceuticals Corp. Method for treating b cell regulated autoimmune disorders
WO2006128129A2 (en) 2005-05-26 2006-11-30 Synta Pharmaceuticals Corp. Method for treating cancer
WO2007023186A1 (en) 2005-08-26 2007-03-01 Laboratoires Serono S.A. Pyrazine derivatives and use as pi3k inhibitors
WO2007044729A2 (en) 2005-10-07 2007-04-19 Exelixis, Inc. N- (3-amino-quinoxalin-2-yl) -sulfonamide derivatives and their use as phosphatidylinositol 3-kinase inhibitors
WO2007054556A1 (en) 2005-11-11 2007-05-18 Æterna Zentaris Gmbh Novel pyridopyrazines and their use as modulators of kinases

Non-Patent Citations (128)

* Cited by examiner, † Cited by third party
Title
"Methoden Der Organischen Chemie (Houben-Weyl)", Dec. 3, 1992, pp. 1-4.
A. Nagel, et al. "Nuclear and Magnetic Resonance Data of Pyrido [2,3-b] Pyrazines and Their Adducts With Amide Ion and Water", Journal of Heterocyclic Chemistry, vol. 16, 1979, pp. 301-304.
Adam R. Renslo, et al. "Self Complimentary Cavitands", Journal of the American Chemical Society, vol. 121, 1999, pp. 7459-7460.
Adindo L. Castelhano, et al. "Glucokinase-Activating Ureas", Bioorganic & Medicinal Shemistry Letters, vol. 15, 2005, pp. 1501-1504.
Alessio Innocenti, et al. "Carbonic Anhydrase Inhibitors: The First On-Resin Screening of a 4- Sulfamoylphenylthiourea Library", Journal of Medicinal Chemistry, vol. 47, 2004, pp. 5224-5229.
Andrew M. Thompson, et al. "3-(3,5-Dimethoxyphenyl)-1,6-Naphthyridine-2,7-Diamines and Related 2- Urea Derivatives Are Potent and Selective Inhibitors of the FGF Receptor-1 Tyrosine Kinase", Journal of Medicinal Chemistry, vol. 43, 2000, pp. 4200-4211.
Andrey Bondev, et al. "Differential Regulation of Lipid and Protein Kinase Activities of Phosphoinositide 3-Kinase In Vitro", Biological Chemistry, vol. 380, Nov. 1999, pp. 1337-1340.
Andrey Bondev, et al. "Differential Regulation of Lipid and Protein Kinase Activities of Phosphoinositide 3-Kinaseγ in Vitro", Biological Chemistry, vol. 380, Nov. 1999, pp. 1337-1340.
Ariamala Gopalsamy, et al. "Pyrazolo[1,5-a] Pyrimidin-7-YL Phenyl Amides As Novel Anti-Proliferative Agents: Parallel Synthesis for Lead Optimization of Amide Region", Bioorganic &Medicinal Chemistry Letters, vol. 15, 2005, pp. 1591-1594.
Arlindo L. Castelhano, et al. "Glucokinase-Activating Ureas", Bioorganic & Medicinal Shemistry Letters, vol. 15, 2005, pp. 1501-1504.
Bart Vanhaesebroeck, et al. "Autophosphorylation of p110Phosphoinositide 3-Kinase: A New Paradigm for the Regulation of Lipid Kinases In Vitro and In Vivo", The EMBO Journal, vol. 18, No. 5, 1999, pp. 1292-1302.
Bart Vanhaesebroeck, et al. "Synthesis and Function of 3-Phosphorylated Inositol Lipids", Annuual Reviews Biochem., vol. 70, 2001, pp. 535-602.
Brader, et al., Tumori 90:2-8 (2004), Phosphoinositide 3-Kinase Signalling Pathways in Tumor Progression, Invasion and Angiogenesis.
Brydon L. Bennett, et al. "SP600125, An Anthrapyrazolone Inhibitor of Jun N-Terminal Kinase", PNAS, vol. 98, No. 24, Nov. 20, 2001, pp. 13681-13686.
Bu-Bing Zeng, et al. "Nitroxyl (HNO) Release From New Functionalized N-Hydroxyurea-Derived Acyl Nitroso-9,10-Dimethylanthracene Cycloadducts", Bioorganic & Medicinal Chemistry Letters, vol. 14, 2004, pp. 5565-5568.
C.L. Leese, et al. "Polyazanaphthalenes. Part I. Some Derivatives of 1:4:5-Triazanaphthalene and Quinoxaline", Japanese Chem. Society,1955, pp. 303-309.
C.M. Atkinson, et al. "Cinnolines and Other Heterocyclic Types in Relation to the Chemotherapy of Trypanosomiasis. Part XI. Some Reactions of Simple Quinoxaline Derivatives", 1955.
Caroline R. Weinstein-Oppenheimer, et al. "The RAF Signal Transduction Cascade as a Target for Chemotherapeutic Intervention in Growth Factor-Responsive Tumors", Pharmacology & Therapeutics, vol. 88, 2000, pp. 229-279.
Carroll Temple, et al. "Antimitotic Agents. Chiral Isomers of Ethyl [5-Amino-1,2-Dihydro-3-(4- Hydroxyhpenyl)-2-Methylpyrido[3,4-b] Pyrazin-7-YL] Carbamate", Journal of Medicinal Chemistry, vol. 35, 1992, pp. 988-993.
Carroll Temple, et al. "Potential Antimitotic Agents. Synthesis of Some Ethyl Benzopyrazin-7- Ylcarbamates, Ethyl Pyrido 3,4-b]pyrazine-7-Ylcarbamates, and Ethyl Pyrido[3,4-e]-As-Triazin-7- Ylcarbamates", Journal of Medicinal Chemistry, vol. 33, 1990, pp. 3044-3050.
Carroll Temple, et al. "Synthesis of Potential Antimalarial Agents. II. 6,8-Disubstituted Pyrido[2,3-b] Pyrazines", Journal of Medicinal Chemistry, vol. 11, Nov. 1968, pp. 1216-1218.
Charles O. Okafor, et al. "Synthesis of Analogues of the 2,3,6,-Triazaphenothiazine Ring System", Journal of Heterocyclic Chemistry, vol. 20, 1983, pp. 199-203.
Chris Marshall, "How Do Small GTPASE Signal Transduction Pathways Regulate Cell Cycle Entry", Current Opinion in Cell Biology, vol. 11, 1999, pp. 732-736.
Corey R. Hopkins, et al. "An Improved Method for the Synthesis of 6-Substituted-5H-Pyrrolo[2,3-b] Pyrazines Via Palladium-Catalyzed Heteroannulation Using Microwave Heating", Tetrahedron Letters, vol. 45, 2004, pp. 8631-8633.
Daniela Catarzi, et al. "Tricyclic Heteroaromatic Systems. Synthesis and A1 and A2a Adenosine Binding Activities of Some 1-Aryl-1, 4-Dihydro-3-Methyl[1]Benzopyrano[2,3-c]Pyrazol-4-Ones, 1-Aryl-4,9-Dihydro-3Methyl-1H-Pyrazolo[3,4-b]Quinolin-4-Ones, and 1-Aryl-1H-IMIDAZO[4,5-b]Quinoxalines", Journal of Medicinal Chemistry, vol. 38, 1995, pp. 1330-1336.
Daniela Catarzi, et al. "Tricyclic Heteroaromatic Systems. Synthesis and A1 and A2a, Adenosine Binding Activities of Some 1-Aryl-1, 4-Dihydro-3-Methyl[1]Benzopyrano[2,3-c]Pyrazol-4-Ones,1-Aryl-4,9-Dihydro-3-Methyl-1H-Pyrazolo[3,4-b]Quinoline-4-Ones, and 1-Aryl-1H-Imidazo[4,5-b]Quinoxalines", Journal of Medicinal Chemistry, vol. 38, 1995, pp. 1330-1336.
Dario R. Alessi, et al. "Mechanism of Activation of Protein Kinase B by Insulin and IGF-1", The EMBO Journal, vol. 15, No. 23, 1996, pp. 6541-6551.
Douglas A. Levin, et al. "Frequent Mutation of the PIK3CA Gene in Ovarian and Breast Cancer", Clinical Cancer Research, vol. 11, No. 8, Apr. 15, 2005, pp. 2875-2878.
Edward C. Taylor, et al. Pterdines. 52. A Convienient Synthesis of 6-Formylpterin, Synthetic Communications, vol. 16, No. 17, 1987, pp. 1865-1868.
Emma Ford, et al. "Regioselective Substitution of 2,3-Dichloro-6-Amino-Quinoxaline", Tetrahedron Letters, vol. 41, 2000, pp. 3197-3198.
Emma R. Parmee, et al. "4-Amino Cyclohexylglycine Analogues As Potent Dipeptidyl Peptidase IV Inhibitors", Bioorganic and Medicinal Chemistry Letters, vol. 14, 2004, pp. 43-46.
Engleman, et al., Nature 7:606 (2006), The Evolution of Phosphatidylinositol 3-Kinases As Regulators of Growth and Matabolism.
Enrico Patrucco, et al. "PI3KModulated the Cardiac Response to Chronic Pressure Overload by Distinct Kinase-Dependent and -Independent Effects", Cell, vol. 118, Aug. 6, 2004, pp. 375-387.
Eugen Muller, et al. "Methoden Der Organischen Chemie (Houben-Weyl)", 1981, pp. 1-11.
F. Chang, et al. "Involvement of PI3K/Akt Pathway in Cell Cycle Progression, Apoptosis, and Neoplastic Transformation: A Target for Cancer Chemotherapy", Leukemia, vol. 17, 2003, pp. 590-603.
F. Chang, et al. "Signal Transduction Mediated by the RAS/RAF/MEK/ERK Pathway From Cytokine Receptors to Transcription Factors: Potential Targeting for Therapeutic Intervention", Leukemia, vol. 17, 2003, pp. 1263-1293.
F. McPhillips, et al. "Association of C-RAF Expression With Survival and Its Targeting With Antisense Oligonucleotides in Ovarian Cancer", British Journal of Cancer, vol. 85, No. 11, 2001, pp. 1753-1758.
Franke, et al., Oncogene 22:8983-8998 (2003), PI3K/AKT and Apoptosis: Size Matters.
Gary A. Molander, et al. "Suzuki-Miyaura Cross-Coupling Reactions of Potassium Alkenyltrifluorocarborates", Journal of Organic Chemistry, vol. 67, No. 24, 2002, pp. 8424-8429.
Gary G. Chiang, et al. "Determination of the Catalytic Activities of Mtor and Other Members of the Phosphoinositide-3-Kinase-Related Kinase Family", Methods in Molecular Biology, vol. 281, pp. 125-141 (2004).
Gary G. Chiang, et al. "Determination of the Catalytic Activities of Mtor and Other Members of the Phosphoinositide-3-Kinase-Related Kinase Family", Methods in Molecular Biology, vol. 281, pp. 125-141.
Gerd Dannhardt, et al. "A Novel Series of 2-Carboxytetrahydroquinolines Provides New Insights Into the Eastern Region of Glycine Site NMDA Antagonists", Arch. Pharm. Pharm. Med. Chem., vol. 333, 2000, pp. 267-274.
Gottfried Heinisch, et at. "Synthesis of N-Aryl-N'-Heteroaryl-Substituted Urea and Thiourea Derivatives and Evaluation of Their Anticonvulsant Activity", Arch. Pharm. Pharm. Med. Chem., vol. 330, 1997, pp. 207-210.
Graham S. Poindexter, et al. "Dihydropyridine Neuropeptide Y Y 1 Receptor Antagonists 2: Bioisoteric Urea Replacements", Bioorganic and Medicinal Chemistry, vol. 12, 2004, pp. 507-521.
Gregory Hughes, et al. "Ethynyl -Extended 2,5-Diphenyl-1,3,4-Oxadiazoles and 2-Phenyl 5-(2-Thienyl)-1,3,4-Oxadiazoles: Synthesis, X-Ray Crystal Structures and Optical Properties", Org. Biomol. Chem., vol. 2, 2004, pp. 3363-3367.
Guichun Yang, et al. "Synthesis of Methyl N-Aryl Oxamate Using Soluble Polymer Support", Synthetic Communications, vol. 36, 2000, pp. 611-619.
H. Friess, et al. "Pancreatic Cancer: The Potential Clinical Relevance of Alterations in Growth Factors and Their Receptors", Journal of Molecular Medicine, vol. 74, 1996, pp. 35-42.
Helen Davies, et al. "Mutations of the Braf Gene in Human Cancer", Letters to Nature, Jun. 9, 2002, pp. 1-6.
Ho Bum Woo, et al. "Synthesis of Novel Curcumin Mimics With Asymmetrical Units and Their Anti-Angiogenic Activity", Bioorganic and Medicinal Chemistry Letters, vol. 15, 2005, pp. 3782-3786.
Hwai Wen Chang, et al. "Transformation of Chicken Cells by the Gene Encoding the Catalytic Subunit of PI 3-Kinase", Science, vol. 276, Jun. 20, 1997, pp. 1848-1850.
Ian G. Campbell, at al. "Mutation of the PIK3CA Gene in Ovarian and Breast Cancer", Cancer Research, vol. 64, Nov. 1, 2004, pp. 7678-7681.
Ian G. Campbell, et al. "Mutation of the PIK3CA Gene in Ovarian and Breast Cancer", Cancer Research, vol. 64, Nov. 1, 2004, pp. 7678-7681.
Jakob Troppmair, et al. "RAF and the Road to Cell Survival: A Tale of Bad Spells, Ring Bearers and Detours", Biochemical Pharmacology, vol. 66, 2003, pp. 1341-1345.
Jaromir Mindl, et al. "Kinetics and Mechanism of Hyrolysis of Aryl N-Methoxycarbamates and Their Derivatives", Collection Czechoslovak Chem. Commun. vol. 48, 1983, pp. 900-905.
Jason W. Szewczyk, et al. "SAR Studies: Designing Potent and Selective LXR Agonists", Bioorganic and Medicinal Chemistry Letters, vol. 16, 2006, pp. 3055-3060.
Jean-Marie Receveur, et al. "4-Acylamino- and 4-Ureidobenzamides as Melanin-Concentrating Hormone (MCH) Receptor 1 Antagonists", Bioorganic and Medicinal Chemistry Letters, vol. 14, 2004, pp. 5075-5080.
Jeffrey A. Engelman et al.; "The evolution of phosphatidylinositol 3-kinases as regulators of growth and metabolism"; Nature Reviews, Genetics, vol. 7, Aug. 2006, pp. 606-619.
Jie-Fei Cheng, et al. "Discovery and Structure-Activity Relationship of Coumarin Derivatives As TNF-α Inhibitors" Bioorganic & Medicinal Chemistry Letters, vol. 14, 2004, pp. 2411-2415.
Jing Chen, et al. "RAF-1 Promotes Cell Survival by Antagonizing Apoptosis Signal-Regulating Kinase 1 Through a MEK-ERK Independent Mechanism", PNAS, vol. 98, No. 14, Jul. 3, 2001, pp. 7783-7788.
Jing Li, et al. "PTEN, A Putative Protein Tyrosine Phosphatase Gene Mutated in Human Brain, Breast, and Prostate Cancer", Science, vol. 275, Mar. 28, 1997.
Jingjun Yin, et al. "PD-Catalyzed N-Arylation of Heteroarylamines", Organic Letters, vol. 4, No. 20, 2002, pp. 3481-3484.
Jiong J. Chen, et al. "Pyrido[2,3-b] Pyrazines From Pyrazine C-Nucleosides: An Unusual Intramolecular Rearrangement", Journal of the American Chemical Society, vol. 118, 1996, pp. 8953-8954.
Joachim K. Seydel, et al. "Synthesis and Quantitative Structure-Activity Relationships of Anticonvulsant 2,3,6-Triaminopyridines", Journal of Medicinal Chemistry, vol. 37, 1994, pp. 3016-3022.
John Mendelsohn, et al. "The EGF Receptor Family as Targets for Cancer Therapy", Oncogene, vol. 19, 2000, pp. 6550-6565.
John W. Huffman, et al. "Enantioselective Synthesis of 1-Methoxy-and 1-Deoxy-2'-Methyl8 -Tetrahydrocannabinols: New Selective Ligands for the CB2 Receptor", Bioorganic & Medicinal Chemistry, vol. 14, 2006, pp. 247-262.
Juan F. Miravet, et al. "Reactive Organogels: Self-Assembled Support for Functional Materials", Organic Letters, vol. 7, No. 22, 2005, pp. 4791-4794.
Judith S. Sebolt-Leopold et al. "Targeting the Mitogen-Activated Protein Kinase Cascade to Treat Cancer", Nature Reviews, vol. 4, Dec. 2004, pp. 937-947.
Kenji Matsuno, et al. "Potent and Selective Inhibitors of PDGF Receptor Phosphorylation. 2. Synthesis, Structure Activity Relationship, Improvement of Aqueous Solubility, and Biological Effects of 4-[4-(Substituted (Thio) Carbamoyl)-1-Piperazinyl]-6,7-Dimethoxyquinazoline Derivatives", Journal of Medicinal Chemistry, vol. 45, No. 2002, pp. 4513-4523.
Khaled All, et al. "Essential Role for the p110beta Phosphoinositide 3-Kinase in the Allergic Response", Letters to Nature, vol. 43, Oct. 21, 2004, pp. 1007-1011.
Khaled All, et al. "Essential Role for the p110β Phosphoinositide 3-Kinase in the Allergic Response", Letters to Nature, vol. 43, Oct. 21, 2004, pp. 1007-1011.
Klaus Okkenhaug, et al. "PI3K in Lymphocyte Development, Differentiation and Activation", Immunology, vol. 3, Apr. 2003, pp. 317-330.
Laleh Shayesteh, et al. "PIK3CA Is Implicated as an Oncogene in Ovarian Cancer", Nature Genetics, vol. 21, Jan. 1999, pp. 99-102.
Lisheng Mao, et al. "Facile Synthesis of 2,3-Distributed Quinoxalines by Suzuki-Miyaura Coupling", Synthesis, No. 15, 2004, pp. 2535-2539.
M. Bohle, et al. "Hetarenes IV. Six Member and Larger Heteroringswith Maximum Unsaturation", Methods of Organic Chemistry(Houben-Weyl), vol. E9, Dec. 18, 1997, pp. 1-8.
M. Sako, "Product Class 20: Pyridopyrazines", Science of Synthesis, vol. 16, No. 20, 2004, pp. 1269- 1290.
M.S.A. El-Gaby, et al. "Some Nucleophilic Reactions With 6-Benzoyl-2,3-Dichloroquinoxaline: Synthesis of Tetrazolo[1,5-a]Quinoxaline, 2-Methylidene-1, 3-Dithiolo[4,5-b] Quinoxalines, Quinoxalino[2,3-b] Quinoxlines and Pyrazolo[1',5':1,2] Imidazolo[4,5-b]-Quinoxalines", Indian Journal of Chemistry, vol. 40B, Mar. 2001, pp. 195-200.
Matthias P. Wymann, et al. "Structure and Function of Phosphoinositide 3-Kinases",Biochimica Et Biophysica Acta, vol. 1436, 1998, pp. 127-150.
Michael A. Crackower, at al. "Regulation of Myocadial Contractility and Cell Size by Distinct PI3K-PTEN Signaling Pathways", Cell, vol. 110, Sep. 20, 2002, pp. 737-749.
Michael A. Crackower, et al. "Regulation of Myocadial Contractility and Cell Size by Distinct PI3K-PTEN Signaling Pathways", Cell, vol. 110, Sep. 20, 2002, pp. 737-749.
Michael R. Myers, et al. "Potent Quinoxaline-Based Inhibitors of PDGF Receptor Tyrosine Kinase Activity. Part 1: SAR Exploration and Effective Bioisosteric Replacement of a Phenyl Substituent", Bioorganic and Medicinal Chemistry Letters, vol. 13, 2003, pp. 3091-3095.
Mircea Darabantu, et al. "Synthesis of New Polyaza Heterocycles. Part 42: Diazines", Tetrahedraon, vol. 61, 2005, pp. 2897-2905.
Natalie A. Dales, et al. "Design and Synthesis of Unsymmetrical Peptidyl Urea Inhibitors of Aspartic Peptidases", Letters vol. 3, No. 15, 2001, pp. 2313-2316.
Ola A. El-Sayed, et al. "Synthesis of Some Novel Quinoline-3-Carboxylic Acids and Pyrimidoquinoline Derivatives As Potential Antimicrobial Agents", Arch. Pharm. Pharm. Med. Chem., vol. 9, 2002, pp. 403-410.
Pablo Rodriguez-Vicana, et al. "Phosphatidylinositol-3-OH Kinase as a Direct Target of RAS", Nature, vol. 70, Aug. 18, 1994, pp. 527-532.
Peter A. Steck, et al. "Identification of a Candidate Tumour Suppressor Gene, MMAC1, at Chromosome 10q23.3 That Is Mutated in Multiple Advanced Cancers", Nature Genetics, vol. 15, Apr. 1997, pp. 356-362.
Pierre Sujobert, et al. "Essential Role for the p110Isoform in Phosphoinositide 3-Kinase Activation and Cell Proliferation in Acute Myeloid Leukemia", Blood, vol. 106, No. 3, Aug. 2005, pp. 1063-1066.
Quingmin Wang et al. "A Convenient Synthesis of Novel N‘-Tert-Butyl-N’-Substituted Benzoyl-N-(Substituted Phenyl) Aminocarbonylhydrazines and Their Derivatives", Synthetic Communications, vol. 34, No. 2, 2004, pp. 255-264.
Rajendra P. Tangallapally, et al. Synthesis and Evaluation of Nitrofuranylamides as Novel Antituberculosis Agents, Journal of Medicinal Chemistry, vol. 47, 2004, pp. 5276-5283.
Rebecca J. Gum, et al. "Acquisition of Sensitivity of Stress-Activated Protein Kinases to the p38 Inhibitor, SB 203580, by Alteration of One or More Amino Acids Within the ATP Binding Pocket", The Journal of Biological Chemistry, vol. 273, No. 25, Jun. 19, 1998, pp. 15605-15610.
Reinhard Wetzker et al.; "Phosphoinositide 3-Kinases as Targets for Therapeutic Intervention"; Curre/It Pharmaceutical Design, 2004,10, pp. 1915-1922.
Reinhard Wetzker, et al. "Phosphoinositide 3-Kinases as Targets for Therapeutic Intervention", Current Pharmaceutical Design, vol. 10, 2004, pp. 1915-1922.
Richard J. Brown, at al. "Synthesis and Properties of Axially-Chiral N-(2,6-Disubstituted) Phenyl Triazolones", Tetrahedron, vol. 60, 2004, pp. 4361-4375.
Richard J. Brown, et al. "Synthesis and Properties of Axially-Chiral N-(2,6-Disubstituted) phenyl Triazolones", Tetrahedron, vol. 60, 2004, pp. 4361-4375.
Rika Hoshino, et al. "Constitutive Activation of the 41-/43-kDa Mitogen-Activated Protein Kinase Signaling Pathway in Human Tumors", Oncogene, vol. 18, 1999, pp. 813-822.
Ritu Dhand, et al. "PI 3-Kinase Is a Dual Specificity Enzyme: Autoregulation by an Intrinsic Protein-Serine Kinase Activity", The EMBO Journal, vol. 13, No. 3, 1994, pp. 522-533.
Robert D. Elliott, et al. "The Isomeric Pyridopyrazines From the Reaction of Some Tetraaminopyridines With Pyruvaldehyde and Benzil", The Journal of Organic Chemistry, vol. 33, No. 6, 1968, pp. 2393-2397.
Robert H. Bradbury, et al. "New Non-Peptide Endothelin-A Receptor Antagonists: Synthesis, Biological Properties, and Structure-Activity Relationships of 5-(Dimethylamino)-N-Pyridyl-, N-pyrimidinyl-, N-Pyridazinyl-, and -N-Pyrazinyl-1-Naphthalenesulfonamides", Journal of Medicinal Chemistry, vol. 40, No. 6, 1997, pp. 996-1004.
Romina Marone et al.; "Targeting phosphoinositide 3-kinase-Moving towards therapy"; Biochimica et Biophysica Acta 1784 (2008), pp. 159-185.
Roy Katso, et al. "Cellular Function of Phosphoinositide 3-Kinases: Implications for Development, Immunity, Homeostasis, and Cancer", Annu. Rev. Cell Dev. Biol., vol. 17, 2001, pp. 615-675.
Rudolph Gotz, et al. "Essential Role of Bag-1 in Differentiation and Survival of Hematopoietic and Neuronal Cells", Nat. Neuroscience, vol. 8, No. 9, Sep. 2005, pp. 1169-1178.
S. Gobec, et al. "Houben-Weyl Methods of Molecular Transformation", Science of Synthesis, vol. 16, 1981, pp. 1-25.
S. Goswami, et al. "Simple and Effiecient Synthesis of 2,7-Difunctionalized-1,8-Naphthyridines", Molecules, vol. 10, 2005, pp. 929-936.
Samir N. Khleif, et al. "A Phase I Vaccine Trial With Peptides Reflecting RAS Oncogene Mutations of Solid Tumors", Journal of Immunotherapy, vol. 22, No. 2, 1999, pp. 155-165.
Sarah M. Moore, et al. "The Presence of a Constitutively Active Phosphoinositide 3-Kinase in Small Cell Lung Cancer Cells Mediates Anchorage-Independent Proliferation Via a Protein Kinase B and p70s6k -Dependent Pathway1 ", Cancer Research, vol. 58, Nov. 15, 1998, pp. 5239-5247.
Satoshi Sasaki, et al. "Discovery of a Thieno[2,3-d] Pyraimidine-2,4-Dione Bearing a p-methoxyureidophenyl Moiety at the 6-Position: A Highly Potent and Orally Bioavailable Non-peptide Antagonist for the Human Luteinizing Hormone-Releasing Hormone Receptor", Journal of Medicinal Chemistry, vol. 46, 2003, pp. 113-124.
Stephen P. Davies, et al. "Specificity and Mechanism of Action of Some Commonly Used Protein Kinase Inhibitors", Biochem. J., vol. 351, 2000, pp. 95-105.
T.S. Osdene, et al. The Synthesis of Compounds With Potential Anti-Folic Acid Activity. Part IV. 3: 6-Diaminopyrido(2:3) Pyrazine., J. Chem. Soc., 1955, pp. 2032-2035.
Takeshi Shiota, et al. "Regioselective Reactions of Organozinc Reagents With 2,4-Dichloroquinoline and 5,7-Dichloropyrazolo[1,5-a] Pyrimidine", Journal of Organic Chemistry, vol. 64, 1999, pp. 453-457.
Teruo Tanaka, et al. "Syntheses of Pyrido[2,3-b]pyrazine Derivatives", Yakugaku Zasshi, vol. 95, No. 9, 1975, pp. 1092-1097 (with partial English Translation).
Thomas Rückle et al.; "PI3inhibition: towards an 'aspirin of the 21st century'?"; Nature Reviews, Drug Discovery vol. 5, Nov. 2006 pp. 903-918.
Timothy S. Lewis, et al. "Signal Transduction Through Map Kinase Cascades", 1998.
Tony Liu, et al. "Mechanistic Studies of LA3+-and ZN2+-Catalyzed Methanolysis of Aryl Phosphate and Phosphorothioate Triesters. Development of Artificial Phosphotriesterase Systems", OBC, vol. 3, 2005, pp. 1525-1533.
Tzvetanka Bondeva, et al. "Bifurcation of Lipid and Protein Kinase Signals of PI3K to the Protein Kinases PKB and MAPK", Science, vol. 282, Oct. 9, 1998, pp. 293-296.
Tzvetanka Bondeva, et al. "Bifurcation of Lipid and Protein Kinase Signals of PI3Kγ to the Protein Kinases PKB and MAPK", Science, vol. 282, Oct. 9, 1998, pp. 293-296.
U.S. Appl. No. 13/523,968, filed Jun. 15, 2012, Claus, et al.
U.S. Appl. No. 13/542,101, filed Jul. 5, 2012, Gerlach, et al.
U.S. Appl. No. 13/770,470, filed Feb. 19, 2013, Claus, et al.
Ulf R. Rapp, et al. "BCL-2 Proteins: Master Switches at the Intersection of Death Signaling and the Survival Control by RAF Kinases", Biochimica Et Biophysica Acta, vol. 1644, 2004, pp. 149-158.
V. Sirivatanauksorn, et al. "Molecular Pattern of Ductal Pancreatic Cancer", Langenbeck's Arch Surg., vol. 383, 1998, pp. 105-115.
Vivanco, et al., Nature Reviews Cancer 2:489-501 (2002), The Phosphatidylinositol 3-Kinase-AKT Pathway in Human Cancer.
W. Mederski, et al. "A General Synthesis of 1-Aryl Carbamoyl-2-Alkyl-4-Aryl Substituted Semicarbazides As Nonbasic Factor Xa Inhibitors", Bioorganic and Medicinal Chemistry Letters, vol. 13 ,2003, pp. 3715-3718.
Wei He, et al. "Potent Quinoxaline-Based Inhibitors of PDGF Receptor Tyrosine Kinase Activity. Part 2: The Synthesis and Biological Activities of RPR127963 an Orally Bioavailable Inhibitor", Bioorganic and Medicinal Chemistry Letters, vol. 13, 2003, pp. 3097-3100.
Xungui He, et al. "Synthesis and Biological Evaluation of Bis and Monocarbonate Prodrugs of 10- Hydroxycamptothecins", Bioorganic and Medicinal Chemistry, vol. 12, 2004, pp. 4003-4008.
Yap et al. "Targeting the P13K-AKT-mTOR pathway:progress, pitfalls, and promises." 2008, Current Opinion in Pharmacology, 8, 393-412. *
Yardena Samuels, et al. "High Frequency of Mutations of the PIK3CA Gene in Human Cancers", Science, vol. 304, Apr. 23, 2004.
Yen-Ying Ma, et al. PIK3CA as an Oncogene in Cervical Cancer, Oncogene, vol. 19, 2000, pp. 2739-2744.
Yiling Lu, et al. "Targeting PI3K-AKT Pathway for Cancer Therapy", Rev. Exp. Clin. Hematol. vol. 7.2, Jun. 2003, pp. 205-228.
Ying Lu, et al. "Design, Synthesis, and Evaluation of 2-Alkoxydihyrocinnamates As PPAR Agonists", Bioorganic & Medicinal Chemistry Letters, vol. 16, 2006, pp. 915-919.

Also Published As

Publication number Publication date
CA2628186A1 (en) 2007-07-19
WO2007079999A2 (en) 2007-07-19
JP2009515854A (en) 2009-04-16
US20070123494A1 (en) 2007-05-31
IL191160A0 (en) 2008-12-29
EP1962854A2 (en) 2008-09-03
IL191160A (en) 2016-03-31
KR20080068116A (en) 2008-07-22
EP1790342A1 (en) 2007-05-30
TW200736253A (en) 2007-10-01
CN101330918B (en) 2015-09-02
JP5448450B2 (en) 2014-03-19
EP1962854B1 (en) 2015-05-06
HK1125823A1 (en) 2009-08-21
KR101473662B1 (en) 2014-12-17
ZA200803867B (en) 2009-05-27
AR056216A1 (en) 2007-09-26
NZ595879A (en) 2013-07-26
PL1962854T3 (en) 2015-10-30
TWI423973B (en) 2014-01-21
ES2543962T3 (en) 2015-08-26
AU2006334721B2 (en) 2011-08-11
UA95464C2 (en) 2011-08-10
BRPI0618451A2 (en) 2011-08-30
WO2007079999A3 (en) 2007-10-04
RU2008123219A (en) 2009-12-20
SG10201401061XA (en) 2014-07-30
RU2487713C2 (en) 2013-07-20
NO20082509L (en) 2008-07-23
CN101330918A (en) 2008-12-24
AU2006334721A1 (en) 2007-07-19

Similar Documents

Publication Publication Date Title
US8937068B2 (en) Pyridopyrazine derivatives and their use
JP6456900B2 (en) Compounds and methods for kinase modulation, and adaptations therefor
JP5726202B2 (en) Novel naphthyridine derivatives and their use as kinase inhibitors
JP5447855B2 (en) Use of novel pyridopyrazine as a kinase modulator
AU2012238678B2 (en) Pyridopyrazine derivatives and their use

Legal Events

Date Code Title Description
AS Assignment

Owner name: ZENTARIS GMBH, GERMANY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:SEIPELT, IRENE;CLAUS, ECKHARD;GUENTHER, ECKHARD;AND OTHERS;SIGNING DATES FROM 20070118 TO 20070124;REEL/FRAME:018873/0738

Owner name: ZENTARIS GMBH, GERMANY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:SEIPELT, IRENE;CLAUS, ECKHARD;GUENTHER, ECKHARD;AND OTHERS;REEL/FRAME:018873/0738;SIGNING DATES FROM 20070118 TO 20070124

FEPP Fee payment procedure

Free format text: PAYOR NUMBER ASSIGNED (ORIGINAL EVENT CODE: ASPN); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

FEPP Fee payment procedure

Free format text: MAINTENANCE FEE REMINDER MAILED (ORIGINAL EVENT CODE: REM.); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

LAPS Lapse for failure to pay maintenance fees

Free format text: PATENT EXPIRED FOR FAILURE TO PAY MAINTENANCE FEES (ORIGINAL EVENT CODE: EXP.); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

STCH Information on status: patent discontinuation

Free format text: PATENT EXPIRED DUE TO NONPAYMENT OF MAINTENANCE FEES UNDER 37 CFR 1.362

FP Lapsed due to failure to pay maintenance fee

Effective date: 20190120